12
12
CANNABIS
                           MEDICAL ASPECTS
No part of this digital document may be reproduced, stored in a retrieval system or transmitted in any form or
by any means. The publisher has taken reasonable care in the preparation of this digital document, but makes no
expressed or implied warranty of any kind and assumes no responsibility for any errors or omissions. No
liability is assumed for incidental or consequential damages in connection with or arising out of information
contained herein. This digital document is sold with the clear understanding that the publisher is not engaged in
rendering legal, medical or any other professional services.
            HEALTH AND HUMAN DEVELOPMENT
                       JOAV MERRICK - SERIES EDITOR
                       NATIONAL INSTITUTE OF CHILD HEALTH
                            AND HUMAN DEVELOPMENT,
                      MINISTRY OF SOCIAL AFFAIRS, JERUSALEM
     CANNABIS
MEDICAL ASPECTS
    BLAIR HENRY
  ARNAV AGARWAL
   EDWARD CHOW
  HATIM A. OMAR
           AND
    JOAV MERRICK
         EDITORS
          New York
Copyright © 2017 by Nova Science Publishers, Inc.
All rights reserved. No part of this book may be reproduced, stored in a retrieval system or
transmitted in any form or by any means: electronic, electrostatic, magnetic, tape, mechanical
photocopying, recording or otherwise without the written permission of the Publisher.
We have partnered with Copyright Clearance Center to make it easy for you to obtain permissions
to reuse content from this publication. Simply navigate to this publication’s page on Nova’s website
and locate the “Get Permission” button below the title description. This button is linked directly to
the title’s permission page on copyright.com. Alternatively, you can visit copyright.com and search
by title, ISBN, or ISSN.
For further questions about using the service on copyright.com, please contact:
Copyright Clearance Center
Phone: +1-(978) 750-8400 Fax: +1-(978) 750-4470          E-mail: info@copyright.com.
Independent verification should be sought for any data, advice or recommendations contained in
this book. In addition, no responsibility is assumed by the publisher for any injury and/or damage
to persons or property arising from any methods, products, instructions, ideas or otherwise contained
in this publication.
This publication is designed to provide accurate and authoritative information with regard to the
subject matter covered herein. It is sold with the clear understanding that the Publisher is not
engaged in rendering legal or any other professional services. If legal or any other expert assistance
is required, the services of a competent person should be sought. FROM A DECLARATION OF
PARTICIPANTS JOINTLY ADOPTED BY A COMMITTEE OF THE AMERICAN BAR
ASSOCIATION AND A COMMITTEE OF PUBLISHERS.
Additional color graphics may be available in the e-book version of this book.
Foreword                                                                        xvii
                Blair Henry, Arnav Agarwal, Edward Chow,
                Hatim A Omar and Joav Merrick
Section one: Introduction                                                         1
Chapter 1       Plants as medical tools                                           3
                Haleh Hashemi, Andrew Hand, Angelique Florentinus-Mefailoski,
                Paul Kerrigan, Phineas Samuel and Jeremy Friedberg
Chapter 2       History of medical cannabis                                      17
                Andrew Hand, Alexia Blake, Paul Kerrigan,
                Phineas Samuel and Jeremy Friedberg
Chapter 3       Cannabis or marijuana                                            27
                Donald E Greydanus and Joav Merrick
Section two: Plant pharmacology                                                  63
Chapter 4       Pharmacology of cannabis                                         65
                Mandakini Sadhir
Chapter 5       The pharmacological properties of cannabis                       71
                Istok Nahtigal, Alexia Blake, Andrew Hand,
                Angelique Florentinus-Mefailoski, Haleh Hashemi Sohi
                and Jeremy Friedberg
Section three: Clinical applications                                             83
Chapter 6       Medical cannabis use in an outpatient palliative care clinic     85
                Noah Spencer, Erynn Shaw and Marissa Slaven
Chapter 7       Four patient perspectives on medical cannabis                    95
                Jeremy Friedberg
xiv                                       Contents
                                           INTRODUCTION
Cannabis has a long history of medicinal use, dating back thousands of years (1). However,
with the discovery of morphine, hypodermic needles and other fast acting synthetic opioids in
the ninetieth and the turn of the twentieth century- cannabis use declined as a medication (2).
    For most of the past six decades, cannabis has been considered a recreational drug, and was
considered illegal in many jurisdictions. Yet in the past few years its association with medicine
has made a dramatic comeback. As illustrated in figure 1, over the past six years (2010-2016)
the terms “medical cannabis” and “medical marijuana” has seen a 4 to 5 fold increase in
references based on a historical trend analysis of the terms noted in the PubMed database. It
    Correspondence: Mr Blair Henry, Senior Ethicist, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue,
       Toronto, ON Canada. E-mail: Blair.Henry@sunnybrook.ca.
xviii                              Haleh Hashemi, Andrew Hand, Angelique Florentinus-Mefailoski et al.
would appear that the term medical cannabis is most often used over medical marijuana, in the
emerging literature.
700
600
                          500
 Frequency of citations
400
200
100
                           0
                            1960              1980            2000            2020
                                            Year of Pulication Search
Figure 1. Number of article titles using the terms: medical cannabis and medical marijuana in PubMed
     In the past several years, claims on the potential for cannabis to treat, cure and prevent a
number of diseases and conditions has led some to query whether these claims are overstated.
     A game changer for medical cannabis has been the ability to consume it without a need to
actually inhale it along with other negative products of combustion. Newer technologies that
allow for the vaporization of the full plant has made it less of a health concern (3).
     Noticeably the evidence on medical cannabis is lacking in both quality and quantity. To
date most of the research has been conducted in Israel, however, with cannabis set to be
legalized for use in Canada (4) and in a recent move in the United States, by the Obama
administration (5), to remove barriers should allow researchers greater access to medical
cannabis for testing in well-designed clinical trials.
     In Canada, the federal marijuana regulations were updated in the summer of 2016 with the
introduction of the Access to Cannabis for Medical Purposes Regulations (ACMPR).
The aim of this newly update regulation (ACMPR) is to treat marijuana like other psychoactive
drugs used for medical purposes. Many of the Colleges (eight provincial) in Canada prohibit or
at best strongly discourage its members from dispensing, providing or accepting delivery of
marijuana for medical purposes (6). Of major concern for most Colleges has been a lack of
good evidence on both medical risks and therapeutic benefits of marijuana- intimating that
                                             Foreword                                                  xix
physicians’ currently prescribing cannabis may be falling short of a fulsome informed consent
process.
     The typical recommendation for Canadian physicians is that medical cannabis should not
be a first line therapy and that documentation should outline that conventional therapies were
attempted but were not successful (6). The legal defense organization for physicians practicing
in Canada- the Canadian Medical Protective Association (CMPA), updated its guidance
document following the passing of ACMPR regulations- a cursory read through this document
quickly identifies a less than positive endorsement for physicians, highlighting mainly potential
risks and limitations to physicians potentially interested in adding medical cannabis to their
treatment recommendations.
     As the medical community has been slow to start- noticeable in the grey literature is the
proliferation of website by patient and advocacy groups making claims that cannabis has the
potential to treat ailments and symptoms ranging from AIDS related illness, Asperger’s,
Bulimia, carpal tunnel syndrome to whiplash- making the differentiation between the
miraculous and mere hype all the more challenging to identify (7-9).
                                           REFERENCES
[1] Borgelt LM, Franson KL, Nussbaum AM, Wang GS. The pharmacologic and clinical effects of medical
    cannabis. Pharmacotherapy 2013;33(2):195–209.
[2] Kritikos PG, Papadaki SP. The history of the poppy and of opium and their expansion in antiquity in the
    eastern Mediterranean area, 1967. URL: https://www.unodc.org/unodc/en/data-and-analysis/bulletin/
    bulletin_1967-01-01_3_page004.html
[3] Grant I, Atkinson JH, Gouaux B, Wilsey B. Medical marijuana: Clearing away the smoke. Open Neurol
    J 2012;6(1):18–25.
[4] Philpott J. Plenary statement for the Honourable Jane Philpott, Minister of Health - UNGASS on the
    World Drug Problem. United Nations General Assembly Special Session on the World Drug Problem.
    URL: http://news.gc.ca/web/article-en.do?nid=1054489
[5] Saint Louis C, Apuzzo M. Obama administration set to remove barrier to marijuana research. New York
    Times 2016 Aug 16 August.
[6] Canadian Medical Protective Association. Medical marijuana: Considerations for Canadian
    doctors, 2014. URL: https://www.cmpa-acpm.ca/en/legal-and-regulatory-proceedings/-/asset_publisher/
    a9unChEc2NP9/content/medical-marijuana-new-regulations-new-college-guidance-for-canadian-
    doctors
[7] United Patient’s Group. Illnessess treatable with medical cannabis, 2016. URL: https://
    unitedpatientsgroup.com/
[8] Walia AQ. Twenty medical studies that show cannabis can be a potential cure for cancer, 2013. URL:
    http://www.collective-evolution.com/2013/08/23/20-medical-studies-that-prove-cannabis-can-cure-
    cancer/
[9] Harding A. Medical marijuana, 2013. URL: http://www.webmd.com/pain-management/features/medical-
    marijuana-uses
SECTION ONE: INTRODUCTION
In: Cannabis: Medical Aspects                                           ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                  © 2017 Nova Science Publishers, Inc.
Chapter 1
      Cannabis has been used for centuries for its fiber, food and as medicine. This review
      highlights the history of cannabis, its uses as a medical tool and the active ingredients found
      in this versatile plant. Many pain management pharmaceuticals widely accepted and used
      today, such as opioids and aspirin, contain plant derived extracts. The evolving cannabis
      story is paralleled to the history of current plant extracts used as pharmaceuticals. Usage,
      side effects and mortality rates of current pain medications are compared to cannabis and
      reveal great potential for cannabis as a safe and effective alternative in pain management.
                                         INTRODUCTION
One of the primary sources of difficulty for doctors to explore the use of cannabis as a medical
tool stems from the idea that they are prescribing a “plant” and not an individual compound.
Although this plant contains a predominant family of active ingredients, the cannabinoids, it is
still a mixture of all the components in the plant tissue or plant extract. To compound this
apprehension, this particular plant has had long and sorted cultural and social-political history
in western civilization that is slowly, yet with difficulty, on the path to a resolution. However,
in the annals of western medicine, the story of this plant’s journey is not new or unique and
there is much to learn from the journey other plants have gone through from obscurity to
common use. The purpose of this chapter is to chronicle two historical plant’s journeys in
modern medicine and what comparisons and differences can be drawn to the story of cannabis.
*
    Correspondence: Jeremy Friedberg, PhD, MedReleaf Corp, Markham Industrial Park, POBox 3040, Markham,
       Ontario, L3R 6C4, Canada. E-mail: jfriedberg@medreleaf.com.
4           Haleh Hashemi, Andrew Hand, Angelique Florentinus-Mefailoski et al.
Secondary metabolites, also referred to as natural products (NP), are organic compounds that
are not directly involved in the natural growth, development, or reproduction of an organism,
and typically result from the activities of biosynthetic pathways. The vast biodiversity of earth’s
flora and fauna have been a tremendous and variable source of useful and medically relevant
compounds and in many cases compounds that cannot be synthesized in vitro (1). The
mechanism by which an organism synthesizes secondary metabolites is often found to be
unique to each organism or it is an expression of the individuality of a species. They are
produced for different reasons from a result of the organism’s adapting to its external
environment, to acting as a possible defense mechanism against predators, or simply in assisting
in the survival of the organism (2, 3).
Medicinal plants
Plants and their extracts have been used as medicinal compounds for thousands of years. Their
unique properties are the result of their evolution. This has resulted in the production of unique
and structurally diverse secondary metabolites. These unique pharmacological properties and
their application by different cultures and regions made them great candidates for new drug
discovery research (4). According to the World Health Organization (WHO), 80% of people
still rely on traditional plant-based medicine for primary health care and 80% of plant derived
drugs were related to their historical application(5). In recent years, advancements in molecular
biology in association with traditional medicine has promoted further investigations and
yielded new drug candidates for the pharmaceutical market (6).
In 2014, the global market for plant-derived drugs was valued at $23.2 billion. It is expected
that this market will reach $35.4 billion by 2020, representing a significant share of the
global pharmaceutical market (8). This increase is a result of (1) the interest expressed by
pharmaceutical companies in new and lower price drugs especially for psychosomatic,
metabolic, and minor disorders and (2) the tendency of people to use modern traditional
medicine. Traditional medicine has been widely used in different types of medication, dietary
products and nutritional supplements since ancient times. Many of them currently are registered
pharmaceuticals through regulatory offices such as the Food and Drug Agency (FDA) once
they surpass clinical trials and demonstrate efficacy and safety (6, 8, 9). To date, 60,000 species
of plants have been screened to yield the 135 known drugs. Considering the number of
unscreened plant species, (approximately >300,000) there is a potential to find 540–653 new
drug candidates in the years to come (10).
Table 1. (Continued)
Cannabidiol (CBD)
bioactive agents has been shown in table 1. A list of plant-derived products that has been used
in clinical tials are shown in table 2.
would be treated with a preparation including the leaves or bark of the willow tree, Salix alba,
which alleviated inflammation, fever, and pain.
    To test historical observations, scientific validation is needed to confirm true relationships.
In 1763, the first scientific description of Salix alba as a treatment for malarial fever in 50
patients is performed by Reverend Edward Stone (27). At the end of his account, Stone states
his hopes; “that it (Salix alba bark powder) may have a fair and full trial in all its variety of
circumstances and situations, and that the world may reap the benefits accruing from it” (27).
    Advances in organic chemistry in the 1820s allowed for the isolation of Salicin from
willow bark (28). Salicin is used successfully to treat rheumatic fever, notably by TJ Maclagan
and Sir William Osler until the end of the 19th century (29).
    In 1838, salicylic acid was derived from Salicin. Pharmaceutical chemists began to
investigate the useful derivatives of salicylic acid which reduced such side effects as
gastrointestinal irritation, resulting in over a dozen such compounds being synthesized by 1908
(26). In 1897, acetylsalicylic acid is synthesized in pure form and by 1899 is being sold
worldwide as Aspirin by Bayer. Current worldwide production of acetylsalicylic acid is
estimated to be 40,000 tons a year, and the number of clinical trials involving the drug is
estimated to be 700-1000 annually (30). Certainly, the powdered bark of Salix alba has had its
advantages and many people continue to reap benefit from its acetylated and pure cousin,
acetylsalicylic acid.
medicine than it could have been in the past. The main method used to determine the strength
of a dose during the late 1800s was to administer cannabis to animals and observe the reaction
(33). Today, laboratory analysis can be far more precise in measurement, using modern
chemical analysis techniques to determine the exact chemical composition of the plant. By
providing this information to physicians it is now possible for them to gauge the dose they are
prescribing the patient (34). Another cause of inconsistency was that cannabis was most likely
obtained from many sources growing different plants (32). Even if the source was the same,
each plant would have a different potency due to genetic variation between seeds and as a
results, acquiring a supply of cannabis that was consistent in composition was nearly impossible
(35). Today this issue can be addressed through the application of clonal propagation and
growing many daughter plants using prunings from a single mother plant. The advantage of
this method is that all daughter plants will be genetically identical and so patients can continue
to have access to consistent and identical medication (36).
     In light of these developments and based off growing evidence of the medical application
of Cannabis, the perception of cannabis is once again changing. We are seeing a general trend
towards cannabis once again becoming a part of western medicine as evidenced by its legal
status for medicinal use in Canada (37). Use of Cannabis as a medicine should be re-evaluated
using modern approaches of science and medicine to determine its true value as a therapeutic
agent.
Medication Utility Common drugs Target Physical dependance Side effects Mortality rate
Cannabis          Cancer, HIV                                    Cannabis Sativa   Endocannabinoid receptor   Rare                  Cognitive and memory impairment                                                                                            Not reported
                  Neurological disorders                                                                                            Increased heart rate, fluctuation in blood pressure. Rare: Stroke, heart infarct
                  Immunity                                                                                                          Anxiety, panic, depression
                  Mood and behaviour                                                                                                Suppressed immune system, growth disorders, apathy, mood/personality changes, hormonal changes
                  Appetite and metabolism
Opioids           Pain (dental, injury, surgery)                 Morphine          Opioid receptor            Yes                   Addiction, brain damage, death                                                                                             <28,000 (9 per 100,000 persons) in USA in 2014 [8]
                  Cough, serious diarrhea                        Codeine                                                            Weakened immune systetm, hallucinations, coma, breathing problems
                  Neuropathy                                     Oxycodone                                                          Sedation, anxiety, hormonal inbalance
                  Sickle cell disease                            Tramadol
                  Headache, migrane                              Fentanyl
                  Fybromyalgia                                   Methadone
                  Anxiety                                        Hydromorphone
                  Cancer
Corticosteroids   Cancer                                         Dexamethasone     Steroid hormone receptor   Yes                   Liver disease, infertility, heart attack, high blood pressure, cancer, depression, psychiatric effects, damage to gonads   Prednisone: 666 of 5626 RA patients in USA over 25 years [4]
                  Arthritis                                      Cortisone                                                          Insomnia, muscle weakness, suppress immune system
                                                                 Hydrocortisone                                                     Glaucoma, osteoporosis
                                                                 Prednisone
Anti-convulsant   Seizures                                       Carbamazepine     GABA receptor              Yes                   Dizziness, suicidal thoughts/actions, depression                                                                           Carbamazepine: 16 per 100,000 cases per year [7]
                  Neuropathy                                     Ethosuximide      PPAR receptor                                    Liver problems, kidney disease, drug interactions, fatigue, drowsiness, nausea, rash, tremor, weight gain                  Gabapentin: 14 of 725 cases in France 1995-2009 [6]
                  Personality disorders                          Gabapentin                                                         Heart disease, depression, confusion, anxiety, constipation, mania
                  Mood
                  Brain disorders (bipolar, mania, depression)
                  Fybromyalgia
                  Insomnia
14           Haleh Hashemi, Andrew Hand, Angelique Florentinus-Mefailoski et al.
    In the United States, 47,000 people died of drug overdose in 2014 and 61% of deaths
associated with drug overdose involved prescription opioids of which oxycodone and
hydrocodone being the most frequently prescribed (45-47). Every year, the number of people
dying from drug poisoning increases exponentially. There is a large demand for an effective
and more safe alternative for relief pain.
    Cannabis has been used for centuries to treat pain, neurological disorders, immunity,
metabolism, mood and behavioural disorders (see table 3) (48, 49). Side effects of Cannabis
use are often mild and reversible with low acute toxicity and no recorded deaths by the Centers
for Disease Control and Prevention (50). Common side effects of cannabis use include euphoria
and temporary cognitive and memory impairment. Hypotension might cause a risk for patients
with cardiovascular diseases (51). In recent studies, data was obtained from patients using
medical cannabis and/or prescription pain medication (PPM) for pain management. Patients
using PPM found medical cannabis to be more efficient in controlling pain than PPM. Patients
on PPM used medical cannabis to significantly reduce opioid intake and improved their quality
of life (52). Overall, cannabis offers a low risk profile and effective alternative in pain
management.
                                          CONCLUSION
The medical cultural apprehension surrounding utility of Cannabis in clinical practice is
understandable. In comparison, the parallels to the stories of plant extracts becoming
pharmaceuticals, and the benefits and associated risks, explains the state of cannabis acceptance
today. Many of these stories have evolved without controversy, while others have been fraught
with social and cultural challenges as well as significant health risks. However, in the case of
cannabis and in comparison to the story of opioids, history is showing us a reasonable and
feasible path forward. The difference, however, in the cannabis story is that the final clinical
utility resides in the plant itself and not just a single compound. Cannabis will continue to be a
viable and beneficial clinical tool, but with ongoing education and research, there is great
potential for further discoveries to the application of cannabis in medicine.
                                           REFERENCES
[1]   Mishra BB, Tiwari VK. Natural products: an evolving role in future drug discovery. Eur J Med Chem
      2011;46:4769–807.
[2]   Kinghorn AD, Pan L, Fletcher JN, Chai H. The relevance of higher plants in lead compound discovery
      programs. J Nat Prod 2011;74(6):1539-55.
[3]   Dias DA, Urban S, Roessner U. A historical overview of natural products in drug discovery. Metabolites
      2012;2(2):303-36.
[4]   Koehn FE, Carter GT. The evolving role of the natural products in drug discovery. Nat Rev Drug Discov
      2005;4(3):206-20.
[5]   Farnsworth NR, Akerele RO, Bingel AS, Soejarto DD, Guo Z. Medicinal plants in therapy. Bull World
      Health Organ 1985;63:965–81.
[6]   David B, Wolfender JL, Dias DA. The pharmaceutical industry and natural products: historical status
      and new trends. Phytochem Rev 2015;14(2):299-315.
                                       Plants as medical tools                                            15
[7]    Cragg GM, Newman DJ. Biodiversity: a continuing source of novel drug leads. Pure Appl Chem
       2005;77:7–24.
[8]    BCC Research. Botanical and plant-derived drugs: global markets. Wellesley, MA: BCC Research,
       2015.
[9]    Naoghare PK, Song JM. Chip-based high throughput screening of herbal medicines. Comb Chem High
       Throughput Screen 2010;13:923–31.
[10]   Schmidt B, Ribnicky DM, Poulev A, Logendra S, Cefalu WT, Raskin I. A natural history of botanical
       therapeutics. Metabolism 2008;57(7 Suppl 1): S3–9.
[11]   Cabral GA, Rogers TJ, Lichtman AH. Turning over a new leaf: Cannabinoid and endocannabinoid
       modulation of immune function. J Neuroimmune Pharmacol 2015;10(2):193-203.
[12]   Gilbert N. Chinese herbal medicine breaks into EU market. Nature News Blog 2012. URL:
       http://blogs.nature.com/news/ 2012/04/Chinese-herbal-medicine-breaks-into-EU-market.
[13]   Bernath J. Pharmaceutical plants. In: Godollo FG, ed. Cultivated plants, primarily as food source,
       Volume 2. Hungary: EOLSS Publishers, 2009.
[14]   Atanasov AG, Waltenberger B, Pferschy-Wenzig EM, Linder T, Wawrosch C, Uhrin P, et al. Discovery
       and resupply of pharmacologically active plant derived natural products: a review. Biotechnol Adv
       2015;33(8):1582–614.
[15]   Rosenblum A, Marsch LA, Joseph H, Portenoy RK. Opioids and the treatment of chronic pain:
       controversies, current status, and future directions. Exp Clin Psychopharmacol 2008;16:405-16.
[16]   Brownstein MJ. A brief history of opiates, opioid peptides, and opioid receptors. Proc Natl Acad Sci
       USA 1993;90:5391-3.
[17]   Kritikos PG, Papadaki SP. The history of the poppy and of opium and their expansion in antiquity in
       the eastern Mediterranean area. United Nations Office on Drugs and Crime 1967. URL: https://www.
       unodc.org/unodc/en/data-and-analysis/bulletin/bulletin_1967-01-01_4_page003.html.
[18]   Dwarakanath C. Use of opium and cannabis in the traditional systems of medicine in India. Bull Narc
       1965;17:15-9.
[19]   Serturner FW. Darstellung der reinen Mohnsäure (Opiumsäure) nebst einer chemischen Untersuchung
       des Opiums mit vorzüglicher Hinsicht auf einen darin neu entdeckten Stoff und die dahin gehörigen
       Bemerkungen. J Pharm F Arzte Apoth Chem 1806;14:47-93. [German]
[20]   Ferrari R, Capraro M, Visentin M. Risk factors in opioid treatment of chronic non-cancer pain: a
       multidisciplinary assessment. In: Racz GB, Noe CE, eds. Pain management: Current issues and
       opinions. Riejeka, Croatia: InTech, 2012:419-58.
[21]   Casey E. History of drug use and drug users in the United States. National Drug Abuse Center
       for Training Resource and Development 1978. URL: http://www.druglibrary.org/schaffer/history/
       casey1.htm.
[22]   Kirsh KL, Vice AK, Passik SD. History of opioids and opiophobia. In: Smith H, Passik S, eds. Pain and
       chemical dependency. Oxford: Oxford University Press, 2008:3-8.
[23]   Janecka A, Fichna J, Janecki T. Opioid receptors and their ligands. Curr Top Med Chem 2004;4:1-17.
[24]   Martin WR. History and development of mixed opioid agonists, partial agonists and antagonists. Br J
       Clin Pharmacol 1979;7:273-9.
[25]   Vadivelu N, Mitra S, Hines RL. Peripheral opioid receptor agonists for analgesia: a comprehensive
       review. J Opioid Manag 2011;7:55-68.
[26]   Jack DB. One hundred years of aspirin. Lancet 1997;350:437–39.
[27]   Stone E. An account of the success of the bark of the willow in the cure of agues. Phil Trans R Soc B
       1763;53:195–200.
[28]   Miner J, Hoffhines A. The discovery of aspirin’s antithrombotic effects. Tex Heart I J 2007;34:179–86.
[29]   Buchanan WW, Kean WF. The treatment of acute rheumatism by salicin, by TJ Maclagan--The Lancet,
       1876. J Rheumatol 2002;29:1321–23.
[30]   Campbell CL, Smyth S, Montescot G, Steinhubl SR. Aspirin dose for the prevention of cardiovascular
       disease, a systematic review. JAMA 2007;297(18):2018–24.
[31]   Sajous CE, Sajous LT. Analytic encyclopedia of practical medicine. Philadelphia,PA: Davis, 1924.
[32]   Mikuriya TH. Marijuana in medicine: past, present and future. Calif Med 1969;110:34-40.
[33]   Zuardi AW. History of cannabis as a medicine: a review. Rev Bras Psiquiatr 2006;28:153-7.
16            Haleh Hashemi, Andrew Hand, Angelique Florentinus-Mefailoski et al.
[34]   De Backer B, Debrus B. Innovative development and validation of an HPLC/DAD method for the
       qualitative and quantitative determination of major cannabinoids in cannabis plant material. J
       Chromatogr B Analyt Technol Biomed Life Sci 2009;877:4115–24.
[35]   Datwyler SL, Weiblen GD. Genetic variation in hemp and marijuana (cannabis sativa l.) according to
       amplified fragment length polymorphisms. J Forensic Sci 2006;51:371-5.
[36]   Coffman CB, Gentner WA, Greenhouse propagation of cannabis sativa l. by vegetative cuttings. Econ
       Bot 1979;33:124-7.
[37]   Fischer B, Kuganesan S, Room R. Medical marijuana programs: implications for cannabis control policy
       – observations from Canada. Int J Drug Policy 2015;26:15-9.
[38]   Snyder MJ, Gibbs LM, Lindsay TJ. Treating painful diabetic peripheral neuropathy: an update. Am Fam
       Physician 2016;94(3):227-34.
[39]   Vargovich AM, McNeil DW, Foley KP, Sperry J. Impact of a psychological opioid-risk evaluation on
       opioid prescribing in primary care. Fam Med 2016;48(7):538-45.
[40]   Peck KR, Ehrentraut JH, Anghelescu DL. Risk factors for opioid misuse in adolescents and young adults
       with focus on oncology setting. J Opioid Manag 2016;12(3):205-16.
[41]   Chester WM, Dasgupta A, Ilse SG, Fries JF, Ward MM. Prednisone use and risk of mortality in patients
       with rheumatoid arthritis: moderation by use of disease-modifying antirheumatic drugs. Arthritis Care
       Res (Hoboken) 2016;68(5):706-10.
[42]   Bernardo WM, Aires FT, de Sa FP. Effectiveness of the association of dexamethasone with antibiotic
       therapy in pediatric patients with bacterial meningitis. Rev Assoc Med Bras 2012;58(3):319-322.
[43]   Fuzier R, Serres I, Guitton E, Lapeyre-Mestre M, Montastruc JL. Adverse drug reactions to gabapentin
       and pregabalin. Drug Saf 2013;36(1):55-62.
[44]   Shorvon S. The antiepileptic drugs. In: Shorvon S. Handbook of epilepsy treatment, 3rd edition.
       Chichester, West Sussex: Wiley-Blackwell, 2010:158-286.
[45]   Rudd RA, Aleshire N, Zibbell JE, Gladden RM. Increases in drug and opioid overdose deaths — United
       States, 2000–2014. MMWR 2016;64(50-51):1378-82.
[46]   Soleimanpour H, Safari S, Shahsavari Nia K, Sanaie S, Alavian SM. Opioid drugs in patients with liver
       disease: a systematic review. Hepat Mon 2016;16(4):e32636.
[47]   Harned M, Sloan P. Safety concerns with long-term opioid use. Expert Opin Drug Saf 2016;15(7):955-
       62.
[48]   Pacher P, Bátkai S, Kunos G. The endocannabinoid system as an emerging target of pharmacotherapy.
       Pharmacol Rev 2006;58(3): 389–462.
[49]   Grotenhermen F. The toxicology of cannabis and cannabis prohibition. Chem Biodivers 2007;4
       (8):1744-69.
[50]   Charlson FJ, Baxter AJ, Dua T, Degenhardt L, Whiteford HA, Vos T. Excess mortality from mental,
       neurological and substance use disorders in the Global Burden of Disease Study 2010. In: Patel V,
       Chisholm D, Dua T, Laxminarayan R, Medina-Mora ME, eds. Mental, neurological, and substance use
       disorders: disease control priorities, 3rd ed (Volume 4). Washington, DC: World Bank, 2016:41-66.
[51]   Perron BE, Bohnert K, Perone AK, Bonn-Miller MO, Ilgen M. Use of prescription pain medications
       among medical cannabis patients: comparisons of pain levels, functioning, and patterns of alcohol and
       Other drug use. J Stud Alcohol Drugs 2015;76(3):406-13.
[52]   Boehnke KF, Litinas E, Clauw DJ. Medical cannabis use is associated with decreased opiate medication
       use in a retrospective cross-sectional survey of patients with chronic pain. J Pain 2016;17(6):739-44.
[53]   Atanas G. Atanasov, Birgit Waltenberger, Eva-Maria Pferschy-Wenzig, Thomas Linderd, Christoph
       Wawroscha, Pavel Uhrine, Veronika Temml, Limei Wang, Stefan Schwaiger, Elke H. Heiss, Judith M.
       Rollinger, Daniela Schuster, Johannes M. Breuss, Valery Bochkov, Marko D. Mihovilovic, Brigitte
       Kopp, Rudolf Bauer, Verena M. Dirsch, and Hermann Stuppner. Discovery and resupply of
       pharmacologically active plant-derived natural products: A review. Biotechnol Adv. 2015 December;
       33(8): 1582–1614.
In: Cannabis: Medical Aspects                                            ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                   © 2017 Nova Science Publishers, Inc.
Chapter 2
       Understanding the cultural and medical history of cannabis use is an important component
       to the successful integration of cannabis in modern clinical practices. This chapter
       chronicles over six thousand years of documented cannabis use in cultural practices,
       medical applications, breeding practices to enhance the pharmacological properties, and
       the various methods by which people have consumed the plant.
                                          INTRODUCTION
Today there is much discussion and debate over cannabis and its use in healthcare. But what is
often left out of the dialogue is the over 6000 years of documented experience people have had
with this plant. Historically, cannabis’ medical applications appear to have been realized by
most cultures, however, it appears that much of our modern day cultural perspectives on
cannabis is based neither on historical evidence nor recent discovery. As with many scientific
disciplines, much can be learned from our collective history. To help with our modern
understanding about cannabis, this chapter provides the reader with a historical account of this
plants’ use, a perspective into the effects of millennia of selective breeding, and insight into the
many ways in which cannabis can be administered.
*
    Corresponding author: Dr. Jeremy Friedberg, MedReleaf Corp., Markham Industrial Park, Markham, Ontario,
       Canada, L3R 6C4, P.O. Box 3040. E-mail: jfriedberg@medreleaf.com.
18                      Andrew Hand, Alexia Blake, Paul Kerrigan et al.
among the ‘five grains’ and was farmed as a major food crop, also playing a major role in the
production of textiles, rope, paper, and oil (2). The first record of its use in medicine comes
from the Pen-ts’ao ching, the world’s oldest pharmacopoeia (3). Although compiled between
0-100 AD, the Pen-ts’ao has been attributed to Emperor Shen-nung, who ruled during 2700
BCE (3). It recognizes cannabis as being useful for over 100 ailments, including rheumatic
pain, gout and malaria (4). The Pen-ts’ao ching also mentions the psychoactive effects of
cannabis stating that “ma-fen (fruit of cannabis), if taken over the long term, it makes one
communicate with spirits and lightens one’s body” (1). Between 117 and 207 AD, Hua T’o, a
physician of the time and the founder of Chinese surgery, described cannabis as an analgesic
(5). He is reported to have used a mixture of cannabis and wine to anesthetize his patients before
surgery (1). As cannabis use increased in China, it spread westward, reaching India by 1000
BCE (2, 3).
     Cannabis spread quickly throughout India and was used extensively, both recreationally
and medicinally (3). It was also adopted and integrated into religious practices, earning mention
in the Atharva Veda, one of the Vedic scriptures of Hinduism, as being among the five sacred
plants of Hinduism, and teaching that a guardian angel lives within its leaves (3). Cannabis is
mentioned within the Vedas as a “source of happiness,” a “joy-giver,” and “bringer of freedom”
(2, 3). The Raja Valabba states that the gods created cannabis out of compassion for humans
(2). In Hinduism, cannabis was smoked during the daily devotional service (2). Due to religious
use in India, it was possible to explore the medicinal benefits of cannabis, which led to the
discovery that cannabis can be used to treat a plethora of diseases and ailments (2). The general
uses in India included use as an analgesic, anticonvulsant, anesthetic, antibiotic, and anti-
inflammatory (3). These properties allowed for the treatment of many diseases, including
epilepsy, rabies, anxiety, rheumatism and even respiratory conditions such as bronchitis and
asthma (3). Cannabis use continued to spread throughout the world and was adopted by many
different cultures (3).
     The Assyrians were aware of cannabis’ psychotropic effects since at least 900 BCE (3). By
450 BCE, cannabis had reached the Mediterranean, as evidenced by a first-hand account from
Herodotus (3). Herodotus writes of a Scythian funeral ceremony, where cannabis seeds were
burned ritually for its euphoric effect (3). In Tibet, cannabis was considered to be sacred, used
extensively in medicine and in Tantric Buddhism to facilitate meditation (3). In Persian
medicine, cannabis’ biphasic effects were clearly noted, emphasizing the distinction between
cannabis’ initial euphoric effects and the dysphoric effects that follow (2). The Persian
physician Avicenna (980 – 1037 AD), one of the most influential medical writers of the
medieval period, published ‘Avicenna’s Canon of Medicine’, a summary of all medical
knowledge of the time (6). His canon was widely studied in western medicine from the
thirteenth to the nineteenth century, having a lasting impact on western medicine (6). Avicenna
recorded cannabis as an effective treatment for gout, edema, infectious wounds and severe
headaches (6). In Arabic medicine, cannabis was regarded as an effective treatment for epilepsy
(7). Recorded first by al-Mayusi, between 900-1000 AD (13), followed by al-Badri, in 1464
AD, who wrote of the chamberlain’s epileptic son who was cured using cannabis leaves (6). In
the 1300s, Arab traders brought cannabis from India to Africa, where it was used to treat
malaria, fever, asthma and dysentery (3). The 1500s saw cannabis reach South America via the
slave trade, which transported Africans along with seeds, from Angola to Brazil (3). In Brazil,
cannabis was used extensively in the African community, including religious rituals such as the
‘Catimbo’ which used cannabis for magical and medical purposes. From Brazil, cannabis
                                History of medical cannabis                                   19
     The prevalence of recreational cannabis use rose significantly in the early 1970s, spiking
from only 5% of people reporting to have used cannabis in 1967, to 44% in 1971 (3). This
massive increase in recreational use brought cannabis into the spotlight, and with the discovery
of cannabis’ active component, Δ9-THC, by Gaoni and Mechoulam in 1964, it became possible
to isolate the principle component, making studying and quantifying its effects possible (3). In
1988, the receptor CB1 was identified (11). It was found to be the binding site of THC, and to
be the most abundant neurotransmitter receptor in the central nervous system (11). This
discovery was followed by the identification of a second cannabinoid receptor, CB2, localized
primarily in the peripheral nervous system and on immune cells (12). The presence of
cannabinoid receptors, concentrated in neural and immune cells, alluded to a possible mode of
action that could be the source of cannabis’ analgesic, sedative and immunoregulatory
properties.
    Many of the genes mentioned above have been sequenced, and molecular markers
detectable via PCR have been developed and validated to correlate with specific chemotypes.
Modern breeders can take advantage of this simple molecular technique in order to expedite
breeding objectives, while using classical breeding techniques in order to select for
other favourable traits, such as yield, disease resistance and flowering time requirements, all
aspects that greatly impact the output of a medical cannabis facility. In the near future more
advanced molecular breeding techniques, such as transgenic gene expression or substitution of
gene promoters with knockdown/overexpression variants could yield dramatically different
chemotypes with potentially novel medical application.
Oral administration
Oral administration through either ingestion or sublingual absorption are also popular methods
of cannabis consumption. Similar to vaporization, oral consumption avoids exposure to smoke
and other hazardous pyrolysis by-products. However, cannabis must be decarboxylated prior
to ingestion.
     Oral administration often involves the consumption of a cannabis extract, rather than the
actual plant material. For oral sprays, such as Sativex, the extract is often mixed with a
diluting/carrier agent, such as propylene glycol (26). Alcohol, flavouring, and sweeteners may
also be added to adjust the viscosity and taste. Application of the product under the tongue
results in rapid absorption due to the high vascularity of the sublingual region. However, taste
is obviously a concern with such products, and a titrated spray dispenser is required for
consistent dosing.
     Alternatively, an infusion can be made by soaking decarboxylated cannabis in butter or
edible oil. This infusion can be used for cooking or baking applications. However, making this
24                       Andrew Hand, Alexia Blake, Paul Kerrigan et al.
infusion is a time consuming and highly-tedious process that, if completed at home, will
produce extracts with unknown and highly variable THC concentrations. Because of this dosing
challenge, capsules may be a safer and more convenient method of cannabis administration,
resulting in higher patient compliance and a lower risk of experiencing adverse effects.
     Despite being a popular historical method of consuming cannabis, tea preparations are not
very popular or recommended for several reasons (27). First, cannabinoid extraction during
steeping will be very low due to the low water solubility of cannabinoids. However, the addition
of cream or non-skim milk may aid in this. Secondly, water temperatures may not be sufficient
to completely decarboxylate cannabinoids. Thirdly, the final concentration of cannabinoids in
the tea will be unknown (and low), making tea a very inefficient way of consuming cannabis.
     Compared to sublingual administration or inhalation, there is a noticeable delay in the onset
of therapeutic action following ingestion (21, 28). For this reason, ingestion may not be a
preferred means of consumption if instant relief is desired.
While inhalation and oral administration are the most common (and therefore the most studied)
methods of cannabis consumption, rectal, transdermal, and ophthalmological administration
are also possible. All of these methods are commonly used for drugs that are not suitable for
oral administration, often due to their potential to irritate the stomach or gastrointestinal tract,
and more commonly due to their low oral bioavailability (21). For cannabis, these methods also
avoid the generation and consumption of smoke and other hazardous combustion by-products.
     Transdermal application may be achieved by incorporating decarboxylated cannabis oil
into topical products, such as lotions, gels, or transdermal patches. Such products may be
most useful for individuals seeking to treat localized, physical pain. Ophthalmological and
suppository products are less common, but animal studies have demonstrated their potential as
alternative methods of cannabis consumption offering rapid absorption (24, 28-30).
                                        CONCLUSION
Cannabis use both culturally and medically has had a long and well documented history.
Cannabis has been used medicinally in many different cultures and upon exposure to western
medicine in the 19th century quickly gained popularity as an analgesic, anticonvulsive, and
hypnotic. These medical properties are innately part of cannabis biology, and over time
selective breeding projects have amplified these traits. The medical properties of this plant
combined with an understanding of the effective methods of consumption help make cannabis
the powerful medication it is today. Much can be learned from this historical record but what
is most salient, is that the use of cannabis to treat clinical symptoms is not new. The challenge
is education, and policy changes to incorporate the nature of cannabis’ atypical consumption
requirements in modern clinical methodology.
                                    History of medical cannabis                                           25
                                     ACKNOWLEDGMENTS
We thank Dean Pelkonen for assistance with graphic design for Figure 1.
                                            REFERENCES
[1]    Hui-Lin L. An archaeological and historical account of cannabis in China. Econ Bot 1974; 28: 437-48.
[2]    Touw M. The religious and medicinal uses of cannabis in China India and Tibet. J Psychoactive Drugs
       1981; 13: 23-34.
[3]    Zuardi AW. History of cannabis as a medicine: a review. Rev Bras Psiquiatr 2006; 28: 153-7.
[4]    Mack A, Joy J. Marijuana as medicine? The science beyond the controversy. Washington: National
       Academy Press, 2001.
[5]    Mikuriya TH. Marijuana in medicine: past, present and future. Calif Med 1969; 110: 34-40.
[6]    Mahdizadeh S, Ghadiri MK, Gorji A. Avicenna's canon of medicine: a review of analgesics and anti-
       inflammatory substances. Avicenna J Phytomed 2015; 5: 182-202.
[7]    Lozano I. The therapeutic use of cannabis sativa (L.) in Arabic medicine. J Cannabis Ther 2001; 1: 63-
       70.
[8]    Whalley B. Cannabis and epilepsy from recreational abuse to therapeutic use. University of Reading
       2007. URL: http://www.medicinalgenomics.com/wp-content/uploads/2013/11/Cannabis_Epilepsy_
       ben-whalley.pdf.
[9]    McMeens RR. Report of the Ohio State Medical Committee on Cannabis Indica. Transactions of the
       Fifteenth Annual Meeting of the Ohio State Medical Society of Ohio White Sulphur Springs 1860. URL:
       http://www.onlinepot.org/medical/Dr_Tods_PDFs/s3_1.pdf.
[10]   Sajous CE, Sajous LT. Analytic cyclopedia of practical medicine. Philadelphia: Davis, 1918.
[11]   Lee MA. The discovery of the endocannabinoid system. The Prop 215 Era 2012. URL: http://www.
       beyondthc.com/wp-content/uploads/2012/07/eCBSystemLee .pdf.
[12]   Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for
       cannabinoids. Nature 1993; 365: 61–5.
[13]   De Meijer EPM, Bagatta M, Carboni A, Crucitti P, Moliterni VMC, Ranallo P, et al. The inheritance of
       chemical phenotype in Cannabis Sativa L. Genetics 2003; 163: 335-46.
[14]   De Meijer EPM, Hammon KM. The inheritance of chemical phenotype in Cannabis sativa L. (II):
       Cannabigerol predominant plants. Euphytica 2005; 145: 189-98.
[15]   Fellermeier M, Eisenreich W, Bacher A, Zenk MH. Biosynthesis of cannabinoids: incorporation
       experiments with 13C-labeled glucoses. FEBS J 2001; 268: 1596-604.
[16]   Rahajo TJ, Chang WT, Choi YH, Peltenburg-Looman AMG, Verpoorte R. Olivetol as product of a
       polyketide synthase in Cannabis sativa L. Plant Sci 2004;166:381-5.
[17]   De Meijer EPM, Hammond KM, Sutton A. The inheritance of chemical phenotype in Cannabis sativa
       L. (IV): cannabinoid-free plants. Euphytica 2009; 168: 95-112.
[18]   De Meijer EPM. The chemical phenotypes (chemotypes) of cannabis. In: Pertwee RG, ed. Handbook of
       cannabis. Oxford: Oxford University Press, 2011: 89-110.
[19]   Fellermeier M, Zenk MH. Prenylation of olivetolate by a hemp transferase yields cannabigerolic acid,
       the precursor of tetrahydrocannabinol. FEBS Lett 1998; 427: 283-5.
[20]   Grotenhermen, Franjo. Pharmacokinetics and pharmacodynamics of cannabinoids. Anesthesiology.
       1997; 86(4): 24–33.
[21]   Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers 2007; 4(8): 1770–804.
[22]   Abramovici H. Information for health care professionals: cannabis (marihuana, marijuana) and the
       cannabinoids. Health Canada 2013. URL: http://www.hc-sc.gc.ca/dhp-mps/alt_formats/pdf/
       marihuana/med/infoprof-eng.pdf.
[23]   Sharma P, Murthy P, Bharath MMS. Chemistry, metabolism, and toxicology of cannabis: Clinical
       implications. Iran J Psychiatry 2012; 7(4): 149–56.
26                         Andrew Hand, Alexia Blake, Paul Kerrigan et al.
[24]   Gieringer DH. Cannabis “vaporization ”: a promising strategy for smoke harm reduction. J Cannabis
       Ther 2001; 14(9): 153–70.
[25]   College of Family Physicians of Canada. Authorizing dried cannabis for chronic pain or anxiety:
       preliminary guidance from the College of Family Physicians of Canada. Mississauga, ON: College of
       Family Physicians of Canada, 2014.
[26]   GW Pharmaceuticals. Product Monograph - Sativex. GW Pharmaceuticals 2015. URL: http:// www.
       gwpharm.com/sativex.aspx.
[27]   Hazekamp A, Bastola K, Rashidi H, Bender J, Verpoorte R. Cannabis tea revisited: a systematic
       evaluation of the cannabinoid composition of cannabis tea. J Ethnopharmacol 2007; 113(1): 85-90.
[28]   Ben Amar M. Cannabinoids in medicine: a review of their therapeutic potential. J Ethnopharmacol 2006;
       105: 1–12.
[29]   Green K, Kearse E, McIntyre O. Interaction between Delta-9-tetrahydrocannabinol and indomethacin.
       Ophthalmic Res 2001; 33(4): 217–20.
[30]   Brenneisen R, Elsohly M. The effect of orally and rectally administered ∆ 9-tetrahydrocannabinol on
       spasticity: a pilot study with 2 patients. Int J Clin Pharmacol Ther 2016; 34(10): 446–52.
In: Cannabis: Medical Aspects                                           ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                  © 2017 Nova Science Publishers, Inc.
Chapter 3
CANNABIS OR MARIJUANA
*   Correspondence: Donald E Greydanus, MD, DrHC (Athens), Professor and Founding Chair, Department of
      Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker MD School of Medicine,
      1000 Oakland Drive, D48G, Kalamazoo, MI 49008-1284, United States. E-mail: Donald.greydanus
      @med.wmich.edu.
28                            Donald E Greydanus and Joav Merrick
                                      INTRODUCTION
A number of chemicals are inhaled for the development of euphoria, including marijuana
(cannabis, pot), methamphetamine, heroin, crack cocaine, phencyclidine, and nitrites (amyl and
butyl) (1). Marijuana (cannabis, pot) has been known to Homo sapiens for thousands of years
and concern has been raised over the past two centuries regarding its potential adverse effects-
--leading to various laws in the United States and other countries to control its production and
use (2). The psychoactive chemical, delta-9-tetrahydrocannabinol (THC) was isolated in the
mid-1960s while other aspects of the endocannabinoid system (cannabinoid receptors [CB1
and CB2] and key endogenous cannabinoids [2-arachidonoyl glycerol and anandamide]) were
identified over 20 years later. Though use of phytocannabinoids are being increasing linked to
improvement of some health conditions, frequent users of cannabis are at increased risk for
adverse effects which can lead to additional health problems (3). Research is identifying
components of cannabis which are not psychoactive and may become established parts of the
pharmacopoeia as the 21st century continues.
The products of cannabis are made from the easily grown hemp plant, Cannabis sativa, and the
psychoactive ingredient, delta-9-tetrahydrocannabinol (THC), is at the heart of the complex
cannabis controversy in this and the past century. The euphoria can last minutes to hours. The
enzyme, Δ1-tetrahydrocannabinolic acid synthase, catalyzes the oxidative cyclization of
cannabigerolic acid (CBGA) into Δ1-tetra-hydrocannabinolic acid, which is the precursor of
THC (4). This enzyme controls the psychoactivity of Cannabis sativa (4) due to THC, which
is present in the C. sativa’s dried leaves, seeds, stems, flowers (sensimilla), and oil (5). The pot
of the 1960s-1970s contained 1-2% THC, while the Hawaiian sensimilla product was 3%; this
is in contrast to current versions with much higher THC percentage as measured by the Potency
Monitoring Project and other research (5, 6). A recent survey in Japan revealed an average
potency of 11.2% with a maximum potency of 22.6% (7). High-potency cannabis is referred to
as “skunk.”
     Cannabis remains a popular global drug that is easily obtained around the world.
Movements are increasing to legalize this drug, because of the research noting positive medical
benefits as well as the intense euphoria it produces along with a popular, though false
impression that this is a “safe” drug (8, 9). Cannabis remains a controversial drug, because its
illegal status places it in conflict with the popular notion that pot is a harmless chemical—a
notion made fashionable or trendy by many media and Hollywood personalities who may
promote its use to enhance their joie de vivre. Such promotions seem to be effective as noted
by the popularity of this illicit drug. However, marijuana (cannabis) has been prohibited in the
United States since the 1937 Marijuana Tax Act as a federal law and is classified by the US
Drug Enforcement Agency (DEA) as an illegal Schedule I drug. The use, sale and possession
of cannabis (marijuana) in the United States is illegal under federal law. However, some states
have created exemptions for medical cannabis use, as well as decriminalized non-medical
cannabis use. In four states, Alaska, Colorado, Oregon and Washington, the sale and possession
                                   Cannabis or marijuana                                        29
of marijuana is legal for both medical and non-medical use, while Washington DC has legalized
personal use, but not commercial sale.
Prevalence
Prevalence data for cannabis can be determined in various ways such as self-report data, waste
water (sewage) analysis, and sales of cigarette paper (10, 11). Research notes that marijuana is
the most commonly used illicit drug on earth (12, 13). The most frequently used substances
among American adolescents are tobacco, alcohol, and marijuana. Marijuana (“weed, pot, hash,
BC Bud, Ganja, grass, smoke, doobs,” others) is an illicit schedule I drug that constitutes about
three-fourths of the illegal drug utilization in the United States (14-16).
    Many studies have been done to confirm the high use of cannabis among adolescents and
young adults of the world. The 2007 European School Survey Project on Alcohol and other
Drugs (ESPAD) reported that lifetime use of cannabis among students (age 15-16 years) in
Europe ranged from 3% in Armenia to 45% in the Czech Republic with an average of 19%
among 35 countries (see table 1). One-third of Canadian university students used cannabis (17).
    The United States Centers for Disease and Prevention (CDC) YRBS (Youth Risk
Behavioral Surveillance) reported a lifetime use (once or more times) among US high school
students of 31.3% in 1991 that increased to 47.2% in 1999 and was 36.8% in 2009 (18). Use of
marijuana by US high schools students that occurred 30 days before the survey ranged from
14.7% in 1991 to 26.7% in 1999 and 20.8% in 2009. Other studies note marijuana use by
adolescents that range from 28% in New York City versus 38% across the United States (19).
Prevalence of cannabis use disorders have increased in veterans in the United States and were
higher in states which allowed cannabis for medical purposes (20).
Marijuana is typically smoked as a joint, but can be taken orally in various foods, teas or
capsules which may be used as “medicinal” marijuana. It can be prepared in food for oral
consumptions, as in brownies, cookies, or spaghetti. Various oral consumptions are found in
different countries. For example, in eastern Iran, there is a special solid “pie” called Majoon
Birjandi, which is consumed by adolescents to reach a cannabis-induced euphoria (21).
     The marijuana (bhang) cigarette is rolled from the C sativa plant (upper leaves, tops, and
stems) that is cut and dried. Hashish refers to dried exudate that comes from the top and
underside of the plant leaves while concentrated hashish distillate is called hashish oil.
Sensimilla is another potent marihuana product that is made from the seedless female flower
of the cannabis plant. The “typical” marijuana cigarette includes about 20 mg of THC that is
produced from a gram of C sativa leaves and buds; however, much variation in potency can be
found (vida supra). THC can be found in the body for up to 2 weeks after use of a single pot
cigarette. A blunt is a cigarette or cigar form made from tobacco and filled with marijuana in a
process called boosting; this can be used to boost the effects of other drugs such as alcohol (22).
30                           Donald E Greydanus and Joav Merrick
                     Armenia: 3%
                     Austria: 17%
                     Belgium (Flanders): 24%
                     Bulgaria: 22%
                     Croatia: 18%
                     Cyrus: 5%
                     Czech Republic: 45%
                     Estonia: 26%
                     Faroe Islands: 6%
                     Finland: 8%
                     France:31%
                     Germany: 20%
                     Greece: 6%
                     Hungary: 13%
                     Iceland: 9%
                     Ireland: 20%
                     Isle of Man: 34%
                     Italy: 23%
                     Latvia: 18%
                     Lithuania: 18%
                     Malta: 13%
                     Monaco: 28%
                     Netherlands: 28%
                     Norway: 6%
                     Poland: 16%
                     Portugal: 13%
                     Romania: 4%
                     Russia: 19%
                     Slovak Republic: 32%
                     Slovenia: 22%
                     Sweden: 7%
                     Switzerland: 33%
                     Ukraine: 14%
                     United Kingdom: 29%
                     AVERAGE: 19%
* Hibell B, Guttormsson U, Ahlström S, Balakireva O, Bjarnason T, Kokkevi A et al. ESPAD (The
    European School Survey Project on Alcohol and Other Drugs) report 2007. Substance use among
    students in 35 European countries. Stockholm, Swedish Council Information Alcohol Other Drugs,
    2009.
Pot is often combined with consumption of alcohol or diazepam which increases cannabis
sedative effects. Addition of various other drugs enhances the euphoric effects. For example,
marijuana is often mixed with various drugs such as nicotine, cocaine, opioids, or hallucinogens
(as LSD [lysergic acid diethylamide]). It is also mixed with drugs such as phencyclidine (PCP)
in which the joint is dipped (hand-rolled) into PCP dissolved in an organic solvent (as
formaldehyde); after drying it is smoked---called “water, wet, or Sherms.” Such additions add
to the complications of pot use. Other additives include glutethimide and methaqualone, which
have been popularized in the past. Persons abusing marijuana who also take disulfiram due to
                                    Cannabis or marijuana                                          31
alcohol abuse can develop increased psychoactive effects of cannabis due to THC blockage by
the disulfiram (23).
      Cannabis users also smoke tobacco and research claimed that this occurs for a variety of
reasons, including shared genetic factors, the similar cue of smoking for both, and withdrawal
symptoms that are seen to some extent in both (24). One study of 467 adults who regularly
smoked both cannabis and tobacco found that one-third started with cannabis first, nearly half
initiated tobacco before cannabis, and most cannabis smokers who stopped tobacco did so after
taking up regular cannabis consumption (25).
      As noted, those who consume marijuana also tend to use other drugs and it may serve as
a gateway drug from early experimentation in adolescence to other drug use (26, 27). For
example, two different studies reported that 45% of college students who illegally used
prescription drugs also abused marijuana, while 24 to 57% also abused alcohol (28). Cannabis
initiation usually follows alcohol use though cannabis use can start before alcohol use and
African-Americans (versus European-Americans) have an increased risk for this cannabis-first
trend (29). Other research notes increased risk for cannabis-related problems in African-
American females versus European-Americans (29). Some research identifies declining
socioeconomic position from childhood to adulthood as a risk factor for cannabis as well as
tobacco consumption (30).
     Some researchers reported that patients seen in a pain clinic were at increased risk for use
of marijuana; for example, one study of these patients with 21, 746 urine specimens found
13.0% incidence of urine with cannabis (tetrahydrocannabinol); also, 4.6% were positive for
cocaine and 1.07% were positive for methamphetamine. (31). A case-crossover research design
study noted that cannabis was a trigger for onset of cocaine use even when genetic influences
and various environmental conditions were held constant (32).
Cannabinoids
Cannabis sativa contains over 60 cannabinoids as well as over 400 other chemicals including
benzopyrene, a known carcinogen. Table 2 lists some of the known cannabinoids (33). The
cannabinoids of the Cannabis sativa plant include cannabidiol (CBD), cannabigerol (CBG), and
cannabinol (CBN) which, in contrast to THC, are not psychoactive. Cannabidiol (CBD) may
activate central nervous system (CNS) limbic and paralimbic regions which can reduce
autonomic arousal and feelings of anxiety; this is in contrast to THC which can be anxiogenic
(34). Cannabigerol is found in higher concentrations in hemp and has been used to lower
intraocular pressure.
     Other research also notes that THC and cannabidiol influence different CNS regions and
thus, have different effects on cannabis users (35). In addition to anxiolytic effects, cannabidiol
has been shown to have anti-emetic, anti-inflammatory, and antipsychotic effects (36). There
is no effect on vital signs (i.e., blood pressure, pulse, body temperature), gastrointestinal transit,
or psychological functioning. Doses up to 1,500 mg per day as well as chronic use of CBD
have been reported as being well tolerated by humans (36). There can be hepatic drug
metabolism inhibition, reduced activities of some drug transporters (i.e., p-glycoprotein), and
lowered capacity of fertilization (36).
32                             Donald E Greydanus and Joav Merrick
    Euphoria is produced from effects of this lipophilic drug on cannabinoid receptors (ECS:
CB 1 and CB 2) in mesocortical and limbic systems; THC also effects the striatum and lateral
prefrontal cortex. Cannabinoid receptors are also found in the liver, gastrointestinal tract,
skeletal tissue, and adipose tissue. There are endogenous ligands for cannabinoid receptors (i.e.,
N-palmitoethanolamide and anandamide) that act like neurotransmitters (26). Part of the
complexity of the cannabis issue is the presence of the endogenous endocannabinoid system
and this system is now reviewed.
Endocannabinoid system
The endocannabinoid system consists of CNS cannabinoid receptors and their endogenous
ligands; ligands (Latin: binding) refers to triggering molecules that bind to a target protein site
(37). Endocannabinoids (endogenous cannabinoid receptor agonsts) include arachidonic acid
derivatives: 2-AG (2-arachidonoyl glycerol) and anandamide (arachidonoyl ethanolamide
[AEA]) (see table 2) (38, 39). AEA and 2-AG are the two most researched endocannabinoids.
The complex effects of this system on emotional and cognitive behavior may be strongly
influenced by various environmental factors (40).
    The endocannabinoid system is identified both as a cause of psychiatric disorders but also
research suggests that proper manipulation of this system may be pharmacologically useful in
management of some psychiatric disorders---such as depression, anxiety, anorexia nervosa, and
others (41). For example, cannabidiol may be beneficial in treatment of psychiatric disorders
(41).
      The endocannabinoid system is involved in processes of brain reward that are related to
drug abuse—as noted in animal and human research; this includes cue-induced relapse of drug
abuse (42). This CNS system is involved in various functions involving memory, emotions,
movement, cell proliferation, and other important cell functions (43). Key neuron classes that
express high levels of CB1 receptors are GABAergic interneurons in such CNS areas as the
cerebral cortex, amygdala, and hippocampus; these areas also contain cholecystokinin, an
important neuropeptide (44).
                                   Cannabis or marijuana                                      33
     The cannabinoid CB (1) receptor (CB1R) is mainly found in the central nervous system
and the CB (2) receptor is expressed in immune system cells and recent research has shown the
importance of this receptor, not only concerning the immune system; both receptors are G-
protein coupled receptors and are involved in adenylate cyclase inhibition (45, 46). The CB1R
is a G-protein coupled receptor that is associated with most of the CNS endocannabinoid
signaling (47). CB1R, also known as CNR1 and CB2R, also known as CNR2 are of importance
and future research will most likely show us new information. It is widely found in the
cerebellum, basal ganglia, and limbic system; the hippocampus has the highest concentration
of cannabinoid receptors. Cannabinoid receptors are found in other tissues including the heart,
lungs, endocrine glands, arteries, immune system, sympathetic ganglia, gastrointestinal tract,
and reproductive tract (38).
     The activation of CB1 receptors can inhibit amino acid and monoamine neurotransmitter
release. Certain lipid derivatives (i.e., 2-arachidonoylglycerol [2-AG] and anandamide [AEA])
can function as endogenous ligands for CB 1 receptors and lead to excitation in such areas as
the cerebellum and hippocampus by inhibition suppression (44). Most drugs of abuse alter brain
levels of endocannabinoids. Since blockade of this system can change the reward behavior
associated with some drugs of abuse, the CNS endocannabinoid system is under active research
to develop medications that may be helpful in treatment of drug abuse—including drug relapse
(42). Cannabis as potential medication (“medicinal marijuana”) is now considered.
Conclusions regarding the “benign” or “malignant” effects of cannabis use influence different
countries’ policies regarding legalization or criminalization of cannabis use (48). This is also
complicated by use of cannabis products in a “medicinal” manner and thus, effects to find the
most “appropriate” variety (varieties) of cannabis that are available (49, 50). A plethora of
medicinal benefits have been identified with marijuana use over past centuries (51-53) (see
table 3).
     In Israel the private company Tikun Olam Ltd was the first, largest and foremost supplier
of medical cannabis in Israel and one of leading medical cannabis companies in the world.
Tikun Olam has been operating under license from the Israel Ministry of Health since 2007. In
2016 medical cannabis is set to hit pharmacy shelves in Israel in the form of cigarettes, cookies
and oil, while the number of doctors permitted to prescribe the natural painkiller and the number
of farmers allowed to grow it will substantially increase, according to a new Health Ministry
reform. At the same time, the entire production and supply chain will be strictly supervised to
ensure that medical cannabis is kept out of the recreational drug market.
     The concern over the addictive and psychomimetic qualities of THC placed active research
on potential medical benefits of this plant on the backburner until recently (53). However,
increased understanding of the cannabinoid signaling system has led to increased research on
potential medicinal uses of cannabis (54); this endocannabinoid system, for example, is being
analyzed for use in treatment of various neuropsychiatric diseases (43).
     Studies are looking at potential benefits of cannabinoids (phytocannabinoids) in
management of neuropathic pain, hypertension, post-stroke neuroprotection, multiple sclerosis,
epilepsy, cancer, and other disorders (55-62). Cancer research, for example, has identified that
cannabinoids can inhibit cancer growth, angiogenesis, and metastasis (62). Cannabinoid-like
34                             Donald E Greydanus and Joav Merrick
anti-inflammatory products may be useful as material for wound dressing due to anti-
inflammatory effects (63). Cannabidiol is under research as a new class of anti-inflammatory
bowel disease drug (64).
A number of designer drugs have become available in the 21st century and cannabis has become
involved in this trend as well. These synthetic cannabinoids (called “legal highs,” “Spice
drugs,” “K2” drugs) are similar to THC found in the C. sativa plant and produce similar effects
to smoking cannabis since they bind to the same cannabinoid brain and peripheral organ
receptors as THC (73-79). These herbal blends have been noted since 2008 in various “herbal”
smoking products sold via the internet and in retail outlets (called “head shops”) that focus on
drug paraphernalia sold for cannabis and other drug use; brand names include such exotic
names as Aroma, Yucatan Fire, Spice Gold, and others (73, 80).
     Though available in some countries, advertised as “safe,” (since they do not resemble the
chemical structure of THC) and considered legal in various locations, they are potentially
dangerous drugs---having up to 10 times the strength of delta-9-THC (74, 81, 82). In a moving
and dangerous cat and mouse game, sellers change the synthetic cannabinoids in attempts to
avoid putting up for sale a specific product identified as illegal in a specific country or area. As
one synthetic cannabinoid is banned, others are produced to take their place as there are over
140 different Spice drugs that are produced (73). They may be marked as some type of
“incense” or “herbal” product and even “air fresheners” but potential adverse reactions remain
(76, 77). Toxicology screens looking at THC may miss the presence of these cannabinoid
designer drugs (76, 78).
     They are typically tobacco and cannabis free but produce similar cannabis effects that can
include withdrawal symptoms, anxiety, intoxication, psychosis, death, and others (vida infra)
(73, 83). Some reports suggest that increased hallucinations and paranoia are noted with these
“spice” synthetic cannabinoids (78). Part of the potential danger is that they can contain various
added but often unknown chemicals that are part of the manufacturing process. However, some
of the products found on the internet do not have significant amounts of impurities and adverse
effects are due to the synthetic cannabinoids themselves and the potential additives (84).
  Addiction (Physiologic)
  Withdrawal syndrome
  Dependence (Psychological) and with heavy use, tolerance
  Variety of negative psychological reactions:
   (anxiety, hallucinations, violent behavior, depression, fear)
  Overt precipitation of psychosis or depression
  Insomnia (can be chronic and improved with trazodone)
  Memory spans that are impaired
  Blunted reflexes
  Flu-like reaction (after stopping this drug after 24 to 60 hours, lasting up to 2 weeks)
  Confusion and cognition impairment
  Alteration of time perception
  Amotivational syndrome (lose interest in school or work success)
  Physiologic responses can include cough, bronchospasm, bronchitis
  Amenorrhea
  Immunologic dysfunction
*Used with permission from Greydanus DE, Feucht CL, Hawver EK. Substance abuse disorders. In:
    Greydanus DE, Patel DR, Omar HA, Feucht C, Merrick J, eds. Adolescent medicine:
    Pharmacotherapeutics in general, mental, and sexual health. Berlin: DeGruyter, 2012:180.
Cannabis hyperemesis
Cannabinoid hyperemesis is noted in some cannabis users who present with usually sudden,
severe, and cyclic (intractable) emesis which resolves with intravenous fluids, antiemetics, and
cannabis cessation. There can be cyclic nausea and abdominal pain as well and after a careful
evaluation, the cause is linked to cannabis use (102). It was first described in Australia in 2004
and may be missed in patients presenting with hyperemesis and abnormal patterns of bathing
(103). Though cannabinoids have been used to treat chronic nausea and emesis, a paradoxical
effect on the gastrointestinal tract in noted in cannabis hyperesis syndrome and three parts are
described: prodromal, hyperemetic, and recovery phases (104).
     The hyperemesis phase is usually resolved in 48 hours. Patients may report temporary
symptomatic improvement with prolonged hot showers or bath exposure and thus, compulsive
hot water bathing has become part of the cannabis (cannabinoid) hyperemesis complex (102,
105-107). Diagnostic confusion with the cyclic vomiting syndrome may occur (104). If
cannabis use resumes, the hyperemesis complex may recur (104, 105, 108).
Pot users tend to have increased risks for dental caries, oral infections, and periodontal disease
(109-111). Dysplastic changes and premalignant lesions can be identified in oral mucosa of
cannabis users (109). Use of local anesthetics in patients intoxicated with cannabis intensifies
and prolongs pot-induced tachycardia (109).
38                           Donald E Greydanus and Joav Merrick
     Exposure to smoking (cannabis or tobacco) leads to contact with many carcinogens (pro-
carcinogens) such as polycyclic aromatic hydrocarbons (112). Cannabis users often smoke
tobacco and drink alcohol which increases carcinogen exposure and risk of oral squamous cell
carcinoma which represent 95% of malignant lesions in the mouth (112).
Pulmonary effects
Some research identifies an anti-inflammatory effect from consumption of the C. sativa plant.
For example, one study of 5,115 adult males that took place over 20 years noted that occasional
and low cumulative cannabis use was not associated with adverse effects on pulmonary
function (113). Murine studies suggest that cannabidiol has an immunosuppressive and anti-
inflammatory effect on acute lung injury because of an increase in extracellular adenosine
(114).
      However, it is known that marijuana, as well as tobacco, contains a toxic combination of
gases and other substances that can be injurious to the pulmonary system (115). Marijuana
smokers usually smoke fewer “joints” than tobacco smokers consume cigarettes; however,
methods of cannabis smoking may place more cannabis particulate matter into the lungs
than noted with typical cigarette smoking (115). Those with cannabis dependence will continue
to use it despite chronic cough, excessive sedation, or other marijuana-related problems.
Combining marijuana with tobacco leads to known tobacco-effects via second-hand smoke.
      Pot use can induce some bronchodilation, but regular or heavy cannabis consumption can
result in generalized airway inflammation with evidence of respiratory epithelial cell injury and
damage to alveolar macrophages which can lead to pulmonary infection (115). Sharing of
cannabis water pipes has led to the development of pulmonary tuberculosis (TB) (116).
Smoking cannabis that contains fungal spores can result in pulmonary aspergillosis in those
with immune-compromised conditions (116, 117).
      There is a dose-related large airway dysfunction with hyperinflation and obstruction of
airflow; one cannabis joint has been noted to be equivalent to 2.5 to 5 cigarettes in terms of this
pulmonary dysfunction (118). Macrophage injury can result in cytokine and nitric oxide
impairment. Smokers of cannabis are typically exposed to more carbon monoxide and tar than
cigarette smokers; this effect is not related to the THC content (119).
      Heavy and/or chronic users of cannabis may have persistent cough, bronchitis, (bullous)
emphysema (chronic obstructive lung disease [COPD]), pulmonary dysplasia, pneumothorax,
tuberculosis (TB), and other respiratory infections (26, 115, 116, 120). Cannabis can lead to
increased airway resistance and large airway inflammation though causal links to COPD or
macroscopic emphysema remain controversial and unproven (92, 93, 118, 121). Smoking both
tobacco and marijuana increases risks for abnormal tracheobronchial histopathology and COPD
(121).
Marijuana smoke contains toxic chemicals in amounts similar to or higher than that found in
tobacco and is linked as a potential respiratory tract carcinogen (26, 93, 96, 120,). Chronic
inflammatory and precancerous airway changes in a dose-dependent relationship as well as
                                   Cannabis or marijuana                                      39
increase in airway cancer are reported in cannabis users (90). Anecdotal reports of upper and
lower respiratory airway cancer have been published (116, 122). For example, a case of small-
cell lung cancer was reported in a 22-year old male who smoked one marijuana joint three times
a week for three years (123). However, specific link of cannabis to lung cancer remains
unproven (92, 93, 124). Current literature suggests that cannabis-only smokers are at lower risk
of lung cancer than tobacco-only smokers (125). However, some epidemiologic data does place
an independent role of cannabis smoking in the development of lung cancer (126, 127).
Cardiovascular effects
Cannabinoids have complex and varying effects on blood pressure depending on which
cannabinoid is being studied (33). Acute effects of cannabis include increase in heart rate along
with an increase (usually mild) in blood pressure and then decreased vascular resistance-
induced orthostatic hypotension (55, 128). Individuals with coronary heart disease may have
increased cardiovascular adverse effects from cannabis use (89). Cannabis-induced ST segment
elevation mimicking the Brugada syndrome has been reported (129, 130). Other drugs, both
licit and illicit (i.e., cocaine), have been linked with the Brugada syndrome as well (131).
      Reported cardiovascular effects linked to cannabis include anecdotal cases of acute
coronary syndrome, congestive heart failure, and arrhythmias (55, 96, 129, 132-135). Patients
with angina may have decreased time with chest pain onset due to the acute effects of cannabis
use; also, myocardial infarction may be triggered by the acute effects of cannabis use (55).
Patients at high risk for coronary heart disease should be advised to avoid using cannabis (55).
      Studies on cannabis also provide evidence of positive or “neutral” effects from cannabis
consumption. For example, though inhalation of marijuana may induce acute coronary
symptoms, ingestion of cannabinoids may have a positive effect on atherosclerotic heart disease
via effects on the endocannabinoid system (134). Also, cannabis use has not been specifically
linked to increased hospitalization due to cardiovascular disease or increased mortality from
cardiovascular etiology (55). If a patient has a cardiac death and has a positive urine test for
cannabinoid, a plasma THC level should also be done before seeking to link the cannabis
history with the cardiac death.
Adolescents and young adults who drive vehicles under the influence of pot (often combined
with alcohol) are at increased risk (two-times) of motor vehicle accidents leading to potential
death and injury (89, 90, 96, 136, 137). Those who consume cannabis without other drugs also
place themselves at increased risk for motor vehicle crashes (137). Individuals driving under
marijuana influence may experience distortion of on-coming vehicle headlights resulting in
motor vehicle crashes. Driving impairment worsens with increasing amounts of cannabis
consumed (138). The problem of driving while under cannabis influence is increasing and in
some areas of California, for example, the rate of nighttime weekend drivers who tested positive
for THC was nearly 20% (139). Unfortunately, tests used in the field to identify if cannabis is
involved in motor vehicle accidents may not be sensitive enough to detect the precise presence
40                           Donald E Greydanus and Joav Merrick
of this drug (140). Cannabis consumption is also involved in non-traffic injuries, especially
falls in the older adult population (141).
Sports doping
Those involved in sports should understand that cannabis is a drug banned by the World Anti-
Doping Agency despite history of pot use in the Olympics and it has been on the list of
prohibited drugs of the International Olympic Committee since 1989 (75, 142). Cannabis
smoking results in reduced exercise test duration during maximal exercising and increased heart
rate at less than maximal exercise levels (55). Cannabis-induced increase in blood pressure and
reduced psychomotor activity can also decrease overall athletic performance. Management of
urine samples from athletes can be problematic because of the intricacies of interpreting urine
samples due to the complexities of prolonged cannabis excretion (143).
Adverse neuropsychological effects of cannabis use must be separated out from the acute
effects of cannabinoids, effects of heavy cannabis consumption, and psychiatric disorders
worsened or even caused by cannabinoids (144). Specific effects in individual cannabis users
are difficult to predict because of the heterogeneity or non-uniformity of different studies that
have been done to seek neuroimaging effects in cannabis use; some authors have concluded it
is difficult to prove major effects of cannabis on brain structure (145, 146). Some research
suggests that identified cannabis-related neurocognitive performance defects disappear after 25
days of cannabis abstinence (147).
     However, other recent research studies are noting that cannabis users demonstrate
important deficits in prospective memory and executive functioning that exist beyond acute
cannabis intoxication (148). The presence of cannabis and nicotine use disorders in parents
appears to increase the risk for major depressive disorder in their late adolescent offspring
(149). Studies in animals and humans suggest a subtle (versus gross) effect on cognitive
functioning with later development of hyperactivity, reduced attention span, impulsivity,
depression, and substance use disorders (150). Most frequently reported adverse effects of
cannabis use include mental slowness, reduced reaction times, and increased anxiety (89).
Dysfunction occurs to dopamine and opioid neurotransmitter systems (151).
     Animal and human research concludes that the developing brain, with its high neuronal
plasticity, is vulnerable to exposure to exogenous cannabinoids, particularly in the
perinatal/prenatal period and during young adolescence (43, 151-154). Animal and human
studies suggest that early onset of cannabis use (i.e., early adolescence) can increase risks for
cognition dysfunction, CNS changes (i.e., low striatal dopamine release), neuropsychiatric
disorders, cannabis dependence, and consumption of additional illict drugs (152, 155).
Persistent or problematic marijuana use can lead to major interference with daily life activities
whether at work, in school, or in one’s home.
                                        Cannabis or marijuana                                41
     Cannabis use often develops in adolescence and early adulthood which, as noted, is a
vulnerable time for subsequent adverse brain effects (47). For example, cannabinoid receptors
are abundant in the CNS white matter in adolescence as well as young adulthood; long-term
cannabis use during this time period can lead to impaired axonal fiber connectivity with
negative effects on the white matter of the brain (156). Cannabis use in early adolescence may
alter CB1R signaling with potentially increased risk for development of psychiatric disorders
(47). Early adolescent cannabis use has been linked with adolescent drop-out behavior in some
research (157).
     Some research links other CNS problems with heavy cannabis use. For example, one study
identified regional brain abnormalities (in the hippocampus and amygdale) in long-term, heavy
pot users—data found in both human and animal studies (158). Animal studies noted
hippocampal-dependent short-term memory deficits that occurred in some but not all rats given
chronic cannabinoid administration (159).
One report of 162 adolescents studied during inpatient management of problems related to drug
dependence (i.e., marijuana, heroin, alcohol, or cocaine abuse) revealed attention deficit
hyperactivity disorder (ADHD) in 34% of them (160). A study from different medical centers
that focused on 600 adolescents (ages 13 to 16 years of age) who were undergoing management
for marijuana-related problems reported that 38% also had ADHD (161). Such studies support
the theory that ADHD and adolescent substance abuse disorder (including cannabis
dependence) can be seen as a developmental disorder with similar underlying physiologic
mechanisms (19).
Cannabis dependence
Psychological dependency and tolerance are classically described in pot smokers. The
American Psychiatric Association’s DSM-IV-TR describes two cannabis use disorders
(cannabis dependence and abuse) and six categories of cannabis-induced disorders (see table
5) (26), while DSM-V has included cannabis withdrawal. Some have placed the dependence
rate at 7-10% of regular users (90), while a susceptibility gene, NRG1, has been associated with
cannabis dependence in African Americans (162).
     Those with cannabis dependence can consume cannabis in potent forms for years and spend
several hours a day in finding as well as taking it; there can be physiologic dependence and also
psychologic dependence (26). Cannabis intoxication may last longer with oral cannabis versus
smoking it. Intoxication may even last up to 24 hours due to effects of enterohepatic circulation
and/or slow release of fat soluable THC and other cannabinoids from fatty tissue. Cannabis use
disorders are more common in males versus females and are most prevalent in the 18 to 30 year
old group (26).
     Depersonalization and derealization episodes are described in pot users (APA, 26).
Anecdotal cases of cannabis-induced depersonalization in adolescents have been reported
(163). A history of conduct disorder in childhood or adolescence and antisocial personality
disorder are risk factors for substance use disorder including cannabis-related disorders (26).
Differentiation of cannabis-induced disorders from primary mental health disorders can be
difficult. Ataxia and aggression are more likely to be seen with phencyclidine (PCP)
intoxication versus cannabis intoxication; aggression with nystagmus or ataxia is more likely
to be from alcohol intoxication (26).
     Demonstrating a direct link between cannabis use and the development of overt depression
has been problematic and not clearly proven (164). Regular cannabis consumption (particularly
daily use) in adolescence has been linked with increased risks for anxiety disorders in
adolescents and young adults, even after the cannabis was stopped (154). Anxiety is linked to
regular or heavy use of cannabis and further research is needed to unravel this connection in
more detail (165). For example anxiety can arise due to fear of being discovered by law
enforcement officials; there can be episodes resembling panic attacks.
Chronic pot users can develop psychological addiction and a withdrawal syndrome comparable
to heroin addiction (8). A heavy marijuana user, whether an adolescent or adult, who suddenly
stops this drug can develop a recognizable withdrawal syndrome as reported by various
research studies (166-172). Withdrawal symptoms can develop within 48 hours of cessation
and include irritability, restlessness, anxiety, aggression, and sleep difficulties. (89).
Withdrawal symptoms tend to subside in 2 to 12 weeks after cannabis abstention (89). As noted,
a CNR1 gene has been linked with abstinence-induced withdrawal symptoms (173). Heavy
cannabis use has been linked with a smaller amygdala and hippocampus while those with CNR1
may be predisposed to smaller hippocampal volume following heavy cannabis use (173). Some
smoke pot to control problems with anger (169-172).
     Withdrawal symptoms are due to the cessation of THC and are relieved with taking delta-
9-THC or simply smoking marijuana (166, 174). Adolescents undergoing treatment for
cannabis dependence experience withdrawal symptoms most acutely during the first week of
cannabis absence and this tends to ease over the next month of abstinence (166, 175). Research
notes that the majority of heavy pot users report several symptoms after stopping cannabis and
this can complicate effective management, especially since cannabis dependent users may
resort to consuming various others drugs to relieve the symptoms of cannabis withdrawal (166,
176). Diagnostic criteria for a formal cannabis withdrawal syndrome (CWS) have been
included in the 5th edition of the American Psychiatric Association’s Diagnostic and Statistical
Manual of Mental Disorders (DSM-V) (177).
                                   Cannabis or marijuana                                        43
Addiction
The identification and isolation of THC in 1964 enhanced specific research into effects of
cannabis on humans and animals (43). Endogenous cannabinoids (particularly CB1 receptor
activation) activate neural mechanisms in the central nervous system (CNS) similar to how
other reward-enhancing drugs induce drug addiction (178-180). This core reward system is part
of the addiction mechanism that use of cannabis develops in the unwary pot smoker as well as
in users of other illict drugs of addiction (181, 182). This mechanism involves the meso-
accumbens reward circuitry of the CNS as well as neural firing of the neurotransmitter,
dopamine. Some research notes that THC can induce striatal effects on dopamine as noted with
other drugs of abuse (183). Though some studies suggest dependence develops faster in cocaine
versus cannabis users, recent research concludes there is no such difference (184).
     Addiction also involves prefrontal cortex (PFC) dysfunction with frontostriatal dysfunction
and the erosion of the ability to stop the addiction; more research is needed to identify potential
impact of cannabinoids in the PFC and its potential role in cannabis dependence (185-187). A
nucleotide polymorphism has been identified in a cannabis receptor-1 gene (CNR1)---
rs2023239-- that is associated with cannabis dependence, cannabis craving, and withdrawal
symptoms due to cannabis abstinence (173). Some neuropeptides called orexins (hypocretins)
originate in the lateral hypothalamus and are linked to features of drug addiction (i.e., cannabis,
nicotine) that include drug craving, relapse, and withdrawal (188).
Chronic use of cannabis, particularly with the newer synthetic cannabis products, is associated
with increased rates of psychosis (189-191). Frequent cannabis use increases the risk by two
times for schizophrenia and psychotic symptoms, perhaps by cannabis-induced disruption of
the endocannabinoid system in which the normal signaling and functioning of this endogenous
system is disturbed (192). Cannabis (marijuana) is commonly used by those with schizophrenia
and can cause paranoia in approximately 40% of persons experimenting with this drug (193).
Patients with schizophrenia who consume cannabis are hospitalized at rates higher than those
who do not use cannabis (194).
     Cannabis-induced schizophrenia is currently theorized as being due to dysfunction of late
postnatal brain maturation in which glutamatergic transmission dysfunction leads to abnormal
prefrontal neurocircuitry (195). Exposure of adolescents to cannabis at certain times in
adolescence and at certain doses may lead to prefrontal cortical circuitry abnormalities with the
result of inducing schizophrenia in susceptible adolescents (195).
     One research group reports a mean time of 7.0 +/- 4.3 years between onset of cannabis use
and onset of psychosis (190). Those at risk for psychosis may be vulnerable to brain volume
loss (i.e., cerebellum, prefrontal cortex, and cingulate) due to use of cannabis (196). Self-
mutilation can also occur with cannabis-induced psychosis (197). Those with psychosis who
use cannabis may not notice improved psychotic symptoms with cessation of their cannabis
(198).
     Most cannabis users do not develop psychosis and this cannabis-psychosis link appears to
be via a complex environmental-genetic-molecular interaction possibly involving anandamide
dysfunction and other biological factors (191, 199, 200). Most research suggests a link between
44                           Donald E Greydanus and Joav Merrick
cannabis use and risk for suicide in patients with psychosis and also those without psychosis
(201). The development of schizophrenia and use of cannabis share a variety of similarities,
including neuropsychological deficits, reduced motivation, hallucinations, and initiation in late
adolescence (202).
    Cannabis is also a very common illicit drug for individuals with psychosis and disruptive
disorders to consume (13, 203). Those with psychosis have a higher rate of cannabis use than
the general population, probably in attempts to utilize the cannabis-induced euphoria to deal
with negative aspects of schizophrenia, such as boredom and depression (12). Cannabis can
also prompt the onset of psychotic symptoms in otherwise healthy people as well, and this
includes paranoia and/or delusional thinking due to effects of THC on striatal and prefrontal
function.
    Thus, use of marijuana can have opposite effects on different users. Some research fails to
find a clear association between use of cannabis and symptoms of psychosis, especially with
low or moderate cannabis use (204). A major component of cannabis, cannabidiol (CBD), has
been shown by some research to have anti-psychotic effects (205). The presence of CBD may
explain the lack of psychosis development in many cannabis users. Studies with cannabidiol
(versus THC) suggest a modulating effect based on functional MRI brain imaging (206).
Schizophrenic patients using cannabis may be particularly sensitive to brain damage from the
cannabis though cannabidiol (CBD) may provide a protective effect from brain volume loss
(202).
                                     MANAGEMENT
Behavioral therapies
The “hedonic” CNS dysregulation seen in drug addiction studies in animals and human subjects
underscores the concept of drug addiction as a brain disease and the necessity of prevention
before this complex brain dysfunction arises (12, 182, 207). Indeed, it is difficult to convince
an addicted cannabis user to stop smoking this plant and thus, prevention via intensive and
comprehensive education is the best option currently available. Unfortunately cannabis
dependence is difficult to treat successfully and few who seek to stop their cannabis addiction
succeed in long-term cessation (180). One of the ironic reasons for this is that chronic cannabis
consumers classically have cognitive impairments which lead to defects in their decision
making skills (208).
      However, there are steps therapists can take in helping this phenomenon of a nation and a
world obsessed with smoking pot. Continued education of the dangers of cannabis should take
place on a persistent and relentless basis. It should be understood that some literature and some
people conclude that the negative impact of smoking cannabis is limited (209). Cannabis
consumers may be cognizant of such literature, which may encourage them to continue to use
this so-called” safe illicit drug. Thus, teaching about the potential problems of cannabis should
always be provided by healthcare providers to help offset this destructive message of “benign”
pot consumption.
      Treatment specifically for cannabis use is needed as management of other or co-existing
drug dependence (such as heroin, tobacco, or alcohol, for example) does not necessarily reduce
                                   Cannabis or marijuana                                        45
striatal control (182). Even brief interventions may have positive benefit in less cannabis use at
3 month follow-up (3) and another study at 12-month follow-up (17).
     Also, therapy seeks to help the addict deal with cannabis craving and relapse triggers, such
as becoming re-exposed to various drugs of addiction; it is also important to help this person
deal with stress and also with finding and avoiding old clues in his/her milieu, such as key
persons, places, or objects (182, 207). Genetic factors also have a role in why some develop
addiction and are recalcitrant to management strategies (207). Involvement of families, schools,
communities, and peer groups are critical in seeking to reduce and prevent cannabis smoking
in adolescents. It is unclear what the interactions are between abuse of illicit drugs and use of
prescription drugs, but the potential drug interactions should be considered in prevention and
treatment plans (225).
                           PHARMACOLOGIC THERAPIES
There are currently no FDA-approved pharmacologic agents for management of cannabis
dependence. Traditionally, pharmacologic agents have not been specifically beneficial in
treating marijuana addiction whether dependence or withdrawal symptoms (226). However, a
careful assessment of each patient is necessary and a reduction in cannabis use can be noted in
some patients under standard pharmacotherapy for mental illness (223). There is no evidence
that one anti-depressant, anxiolytic, or antipsychotic is more effective than another. Cannabis
use disorders are common in those with schizophrenic spectrum disorders; however, there is
no current literature that guides clinicians in the best treatment approaches for this dual
diagnosis (227). Knowledge, however, is slowly emerging to guide pharmacologic therapies
for cannabis-induced problems in the 21st century.
     Individuals with co-occurrence of cannabis and tobacco use tend to have higher abstinence
rates if treatment includes measures aimed at dual abstinence (24). Thus, pharmacotherapy for
nicotine addiction may help the individual stop marijuana use as well by removing use of and
thus, influence of tobacco. Pot-associated persistent insomnia may be improved with use of
trazodone. Research is looking at new methods of treatment that will emerge from the study of
the genetics of addiction including gene-milieu interplay and role of genetic variation (228).
                              CANNABIS INTOXICATION
Pharmacologic management typically centers on use of benzodiazepines or atypical
antipsychotics. Propranolol and rimonabant (vida infra) have been reported to be beneficial in
management of acute, physiologic effects of cannabis intoxication; use of flumazenil and
cannabidiol is under current study in this regard (229).
                               CANNABIS WITHDRAWAL
Oral THC (dronabinol) may relieve cannabis withdrawal symptoms but not relapse (230, 231).
Adding dronabinol and lofexidine (alpha-adrenergic receptor agonist) can lower the severity of
                                   Cannabis or marijuana                                       47
withdrawal and lower relapse rates in those with cannabis dependence (232). The anti-
depressant mirtazapine can also help those undergoing cannabis withdrawal.
                        CANNABIS-ASSOCIATED PSYCHOSIS
Cannabidiol has been shown in animal and human studies to have anti-psychotic effects and it
may become a useful pharmacotherapeutic agent for schizophrenia management (205).
Research has therapeutically targeted the cannabinoid (CB1) receptor since delta-9-THC is a
partial CB1 receptor agonist. Treatment of patients with psychotic disorders and cannabis use
should include seeking to reduce or stop the cannabis use, perhaps using cannabinoid agonist
medication (219). Those with psychosis can be provided with appropriate anti-psychotic
medications.
                               CANNABIS DEPENDENCE
Nabilone is a synthetic THC analogue with improved bioavailability over dronabinol and is
under current research as a potential medication for marijuana dependence as it may lead to a
positive mood and limited adverse cognitive effects in marijuana smokers (233) (see chapter
5). Olanzapine can reduce psychomimetic effects of THC in some individuals and its potential
benefit is under study (234). Rimonabant and the anxiolytic buspirone have provided
some efficacy in cannabis addicts on maintenance therapy. Rimonabant is a CB1-selective
cannabinoid receptor antagonist/inverse agonist that has been under research for obesity
treatment as well as treatment of nicotine and marijuana addiction. It is not available in the
United States and was pulled from the European market in October of 2008 due to a high risk
to benefit ratio; concerns have been identified regarding major psychiatric adverse effects (i.e.,
depression and suicide) as well as overall efficacy (166).
Oxytocin
Animal research notes that various drugs, including cannabis, can induce chronic changes in
markers of oxytocin function with resultant social behavior dysfunction. Oxytocin is a
neurohypophyseal hormone that has been under research in the regulation of drug abuse (235).
This neuropeptide may serve as a neuromodulator on neurotransmission of dopamine in the
nucleus accumbens as well as effects on the hippocampus (235). Central nervous system
oxytocin pathways may offer a way of improving mood and social deficits found in some
individuals with drug addiction (236). Research suggests that use of intranasal oxytocin may
be useful in correcting the negative cannabis (and other illicit drug) effect on social behavior
and perhaps protect the individual from addictive disorders. (237).
NAC
suggested as an adjuvant treatment (1,200 mg per day) along with psychological management
(238).
Others
Research has failed to validate the use of naltrexone for the treatment of cannabis dependence.
Entacapone is a member of the drug class called nitrocatechols that is used in the management
of Parkinson’s disease. It is an inhibitor of COMT (catechol-O-methyltransferase) and is under
research as a possible drug for cannabis dependence.
    Other drugs under research include lithium, dronabinol, URB597 (fatty acid amide
hydrolase [FAAH] inhibitor), methyllycaconitine or MLA (nicotinic alpha-7-receptor
antagonist), and endocannabinoid metabolizing enzymes (239).
                                        CONCLUSION
Research must provide more specific information on how cannabis and products derived from
cannabis can be useful in medical management of illness or if the cannabis risks simply
outweigh any potential benefit (240). Does cannabis have advantages over specific
cannabinoids? Can the psychotropic effects of cannabis be reduced, while utilizing potential
medicinal benefits of CB1 receptors activation? Oral cannabis undergoes changes which
produce a narrower therapeutic window and raises questions regarding whether or not
medicinal cannabis products can be given orally or must be used as a mist or smoke for positive
and optimal benefit. It is difficult to find an oral dose that benefits most without induced
unacceptable or unwanted adverse effects in many unwary consumers (240).
     Generalized consumption of the unprocessed Cannabis sativa plant can lead to
considerable public health risks including increased schizophrenia, psychosis, dependence, and
other risks as noted in this review (241). A medical role for specific cannabinoid compounds
remains under active medical research (242, 243). What is well-known is that there are many
potential medical and psychiatric adverse effects to smoking cannabis and its synthetic
derivatives. Links between use of alcohol, cannabis, and other illicit drugs continues to be
unveiled by research (244). Research is identifying new techniques of cannabis identification
to assist the police and other authorities in forensic investigation (245). Finally, treatment of an
individual with cannabis dependence is very difficult and requires more research in the 21st
century (246, 247). Indeed, Cannabis sativa, a controversial plant known for thousands of
years, remains disputatious and contentious in the 21st century (248).
Medical cannabis
In recent years medical cannabis has been used for patients with cancer, PTSD (post traumatic
stress syndrome), Crohn disease, colitis, Tourette syndrome, Parkinson disease, ALS
(amyotropic lateral sclerosis), MS (multiple sclerosis), epilepsy, Alzheimer disease and AIDS.
                                   Cannabis or marijuana                                       49
                                     ACKNOWLEDGMENTS
This paper is an adapted and revised version of an earlier publication: Greydanus DE, Hawver
EK, Greydanus MM, Merrick J. Marijuana: Current concepts. Front Public Health 2013 Oct
10. doi: 10.3389/fpubh.2013.00042.
                                           REFERENCES
[1]    Tashkin DP. Smoked marijuana as a cause of lung injury. Monaldi Arch Chest Dis 2005;63(2):93-100.
[2]    Anthony JC. Steppingstone and gateway ideals: a discussion of origins, research challenges, and
       promising lines of research for the future. Drug Alcohol Depend 2012 May 7.
[3]    Fischer B, Jones W, Shuper P, Rehm J. 12-month follow-up of an exploratory “brief intervention” for
       high-frequency cannabis use among Canadian university students. Subst Abuse Treat Prev Policy
       2012;7(1):15.
[4]    Shoyama Y, Tamada T, Kurihara K, Takeuchi A, Taura F, Arai S, et al. Structure and function of Δ1-
       tetrahydrocannabinolic acid (THCA) synthase, the enzyme controlling the psychoactivity of Cannabis
       sativa. J Mol Biol 2012 Jul 2.
[5]    Greydanus DE, Hawver EK. Substance use and abuse in adolescents. In: Greydanus DE, Patel DR, Pratt
       HD, Calles Jr JL, Nazeer A, Merrick J, eds. Behavioral Pediatrics, 4 edition. New York: Nova Science,
       2015:497-535.
[6]    Goullé JP, Saussereau E, Lacroix C. Delta-9-tetrahydrocannabinol pharmacokinetics. Ann Pharm Fr
       2008;66(4):232-44.
[7]    Tsumura Y, Aoki R, Tokieda Y, Akutsu M, Kawase Y, Kataoka T, et al. A survey of the potency of
       Japanese illicit cannabis in fiscal year 2010. Forensic Sci Int 2012 May 1.
[8]    Greydanus DE, Patel DR. Substance abuse in the adolescent. In: Greydanus DE, Patel DR, Pratt HD,
       eds. Essential adolescent medicine. New York: McGraw-Hill; 2006:704-5.
[9]    Clark PA, Capuzzi K, Fick C. Medical marijuana: medical necessity versus political agenda. Med Sci
       Mont 2011;17(12):RA249-61.
[10]   Steppan M, Kraus L, Piontek D, Siciliano V. Are cannabis prevalence estimates comparable across
       countries and regions? A cross-cultural validation using search engine query data. Int J Drug Policy
       2012 Jul 16.
[11]   Thomas KV, Bijlsma L, Castiglioni S, Covaci A, Emke E, Grabic R, et al. Comparing illicit drug use in
       19 European cities through sewage analysis. Sci Total Environ 2012 Jul 25.
[12]   Kolliakou A, Fusar-Poli P, Atakan Z. Cannabis abuse and vulnerability to psychosis: targeting
       preventive services. Curr Pharm Des 2012;18(4):542-9.
[13]   Teesson M, Slade T, Swift W, Mills K, Memedovic S, Mewton L, et al. Prevalence, correlates and
       comorbidity of DSM-IV cannabis use and cannabis use disorders in Australia. Aust NZ J Psychiatry
       2012 Sept 14.
[14]   Greydanus DE, Feucht CL, Hawver EK. Substance abuse disorders. In: Greydanus DE, Patel DR, Omar
       HA, Feucht C, Merrick J, eds. Adolescent medicine: Pharmacotherapeutics in general, mental, and
       sexual health. Berlin: De Gruyter, 2012:157-99.
[15]   Greydanus DE, Patel DR. Substance abuse in adolescents: a complex conundrum for the clinician.
       Pediatr Clin North Am 2003;59(5):1179-223.
[16]   Greydanus DE, Patel DR. Substance abuse in adolescents: current concepts. Dis-A-Mon 2005;51
       (7):392-431.
[17]   Fischer B, Dawe M, McGuire F, Shuper PA, Capler R, Bilsker D, et al. Feasibility and impact of brief
       interventions for frequent cannabis users in Canada. J Subst Abuse Treat 2013;44(1):132-8.
[18]   Youth Risk Behavior Surveillance (YRBS)—US 2009. MMWR 2010;59(SS-5):1-142.
                                        Cannabis or marijuana                                               51
[19]   Ivanov I, Pearson A, Kaplan K, Newcorn J. Attention deficit hyperactivity disorder and comorbid
       substance abuse. In: Greydanus DE, Calles Jr JL, Patel DR, Nazeer A, Merrick J. eds. Clinical aspects
       of psychopharmacology in childhood and adolescence. New York: Nova Science, 2011:33-49.
[20]   Bonn-Miller MO, Harris AH, Trafton JA. Prevalence of cannabis use disorders diagnoses among
       veterans in 2002, 2008, and 2009. Psychol Serv 2012 May 7.
[21]   Mehrpour O, Karrari P, Afshari R. Recreational use and overdose of ingested processed cannabis
       (Majoon Birjandi) in the eastern Iran. Hum Exp Toxicol 2012 Jun 29.
[22]   Lipperman-Kreda S, Lee JP. Boost your high: cigarette smoking to enhance alcohol and drug effects
       among Southeast Asian American youth. J Drug Issues 2011;41(4):509-22.
[23]   Greydanus DE, Feucht C, Patel DR. Substance abuse disorders in adolescents: pharmacologic
       management. In: Greydanus DE, Calles Jr JL, Patel DR, Nazeer A, Merrick J, eds. Clinical aspects of
       psychopharmacology in childhood and adolescence. New York: Nova Science, 2011:178.
[24]   Agrawal A, Budney AJ, Lynskey MT. The co-occurring use and misuse of cannabis and tobacco: a
       review. Addiction 2012;107(7):1221-33.
[25]   Simmons MS, Tashkin DP. The relationship of tobacco and marijuana smoking characteristics. Life Sci
       1995;56(23-24):2185-91.
[26]   American Psychiatric Association. Diagnostic and statistical manual of mental disorders, DSM-IV-TR,
       4th ed. Washington, DC: American Psychiatric Association, 2000.
[27]   Olthuis JV, Darredeau C, Barrett SP. Substance use initiation: the role of simultaneous polysubstance
       use. Drug Alcohol Rev 2012 May 21. doi: 10.1111/j.1465-362.2012.00470x.
[28]   Barrett SP, Darredeau C, Pihl RO. Patterns of simultaneous polysubstance use in drug using university
       students. Hum Psychopharmacol 2006;21(4);255-63.
[29]   Sartor CE, Agrawal A, Lynskey MT, Duncan AE, Grant JD, Nelson EC, et al. Cannabis or alcohol first?
       Differences by ethnicity and in risk for rapid progression to cannabis-related problems in women.
       Psychol Med 2013;43(4):813-23.
[30]   Bowes L, Chollet A, Fombonne E, Galéra C, Melchior M. Lifetime SEP and tobacco and cannabis use.
       Eur J Public Health 2013;23(2):322-7.
[31]   Pesce A, West C, Rosenthal M, West R, Crews B, Mikel C, et al. Marijuana correlates with use of other
       illicit drugs in a pain patient population. Pain Physician 2010;13(3):283-7.
[32]   O’Brien MS, Comment LA, Liang KY, Anthony JC. Does cannabis onset trigger cocaine onset? A case
       crossover approach. Int J Methods Psychiatr Res 2012;21(1):66-75.
[33]   Malinowska B, Baranowska-Kuczko M, Schlicker E. Triphasic blood pressure responses to
       cannabinoids: do we understand the mechanism? Br J Pharmacol 2012;165(7):2073-88.
[34]   Fusar-Poli P, Crippa JA, Bhattacharyva S, Borgwardt SJ, Allen P, Martin-Santos R, et al. Distinct effects
       of {delta}9-tetrahydrcannabinol ad cannabidiol on neural activation during emotional processing. Arch
       Gen Psychiatry 2009;66(1):95-105.
[35]   Borgwardt SJ, Allen P, Bhattacharyya S, Fusar-Poli P, Cirppa JA, Seal ML, et al. Neural basis of Delta-
       9-tetrahydrocannabinol and cannabidiol: effects during response inhibition. Biol Psychiatry 2008;64
       (11):966-73.
[36]   Bergamaschi MM, Queiroz RH, Zuardi AW, Crippa JA. Safety and side effects of cannabidiol: a
       cannabis sativa constituent. Curr Drug Saf 2011;6(4):237-49.
[37]   Solinas M, Yasar S, Goldberg SR. Endocannabinoid system involvement in brain reward processes
       related to drug abuse. Pharmacol Res 2007;56(5):393-405.
[38]   Grotenhermen F. Cannabinoids. Curr drug targets CNS Neurol Disord 2005;4(5):507-30.
[39]   Mechoulam R, Parker LA. The endocannabinoid system and the brain. Annu Rev Psychol 2013;64:21-
       47.
[40]   Zanettini C, Panlilio LV, Alicki M, Goldberg SR, Haller J, Yasar S. Effects of endocannabinoid system
       modulation on cognitive and emotional behavior. Front Behav Neurosci 2011;5:57-62.
[41]   Marco EM, Garcia-Gutiérrez MS, Bermúdez-Silva FJ, Moreira FA, Guimarães F, Manzanares J, et al.
       Endocannabinoid system and psychiatry: in search of a neurobiological basis for detrimental and
       potential therapeutic effects. Front Behav Neurosci 2011;55:63-70.
52                               Donald E Greydanus and Joav Merrick
[42]   Fattore L, Spano MS, Deiana S, Melis V, Cossu G, Fadda P, et al. An endocannabinoid mechanism in
       relapse to drug seeking: a review of animal studies and clinical perspectives. Brain Res Rev
       2007;53(1):1-16.
[43]   Trezza V, Cuomo Vo, Vanderschuren LJ. Cannabis and the developing brain: insights from behavior.
       Eur J Pharmacol 2008;585(2-3):441-52.
[44]   Iversen L. Cannabis and the brain. Brain 2003;26(Pt 6):1252-70.
[45]   Marriott KS, Huffman JW. Recent advances in the development of selective ligands for the cannabinoid
       CB (2) receptor. Curr Top Med Chem 2008;8(3):187-204.
[46]   Guindon J, Hohmann AG. The endocannabinoid system and cancer: therapeutic implication. Br J
       Pharmacol 2011;163(7):1447-63.
[47]   Caballero A, Tseng KY. Association of cannabis use during adolescence, prefrontal CB1 receptor
       signaling, and schizophrenia. Front Pharmacol 2012;3:101-4.
[48]   Hall W. The adverse health effects of cannabis use: what are they, and what are their implications for
       policy? Int J Drug Policy 2009;20(6):458-66.
[49]   Hazelkamp A, Fischedick JT. Cannabis-from cultivar to chemovar. Drug Test Anal 2012; doi.
       10.1002/dta.407.
[50]   Cannabis-based medicines—GW pharmaceuticals: high CBD, high THC, medicinal cannabis—GW
       pharmaceuticals, THC:CBD. Drugs R D 2003;4(5):306-9.
[51]   Naftali T, Lev LB, Yablecovitch D, Half E, Konikoff FM. Treatment of Crohn’s disease with cannabis:
       an observational study. Isr Med Assoc J 2011;13(8):455-8.
[52]   Collen M. Prescribing cannabis for harm reduction. Harm Reduct J 2012;9(1):1.
[53]   Fernandez-Ruiz J. Cannabinoid drugs for neurological diseases: what is behind? Rev Neurol
       2012;54(10):613-28.
[54]   Kraft B. Is there any clinically relevant cannabinoid-induced analgesia? Pharmacology 2012;89(5-
       6):237-46.
[55]   Sidney S. Cardiovascular consequences of marijuana use. J Clin Pharmacol 2002;42(11 Suppl):64S-
       70S.
[56]   Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, MUSEC Research Group. Multiple sclerosis
       and extract of cannabis: Results of the MUSEC trial. J Neurol Neurosurg Psychiatry 2012;83(11):1125-
       32.
[57]   Hofmann ME, Frazier CJ. Marijuana, endocannabinoids, and epilepsy: Potential and challenges for
       improved therapeutic intervention. Exp Neurol 2011;233:112-25.
[58]   Calvaruso G, Pellerito O, Notaro A, Giuliano M. Cannabinoid-associated cell death mechanisms in
       tumor models. Int J Oncol 2012L41(2):407-13.
[59]   Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H, et al. Smoked cannabis for
       spasticity in multiple sclerosis: a randomized, placebo-controlled study. CMAJ 2012;184:10.
[60]   Wade D. Evaluation of the safety and tolerability profile of Sativex: is it reassuring enough? Expert Rev
       Neurother 2012;12(4 Suppl):9-14.
[61]   Jones NA, Glyn SE, Akiyama S, Hill TD, Hill AJ, Weston SE, et al. Cannabidiol exerts anti-convulsant
       effects in animal models of temporal lobe and partial seizures. Seizure 2012;21(5):344-52.
[62]   Caffarel MM, Andradas C, Pérez-Gómez E, Guzmán M, Sánchez C. Cannabinoids: a new hope for
       breast cancer therapy? Cancer Treat Rev 2012;38(7):911-8.
[63]   Styrczewska M, Kulma A, Ratajczak K, Amarowicz R, Szope J. Cannabinoid-like anti-inflammatory
       compounds from flax fiber. Cell Mol Biol Lett 2012;17(3):479-99.
[64]   Esposito G, Filippis DD, Cirillo C, Luvone T, Capoccia E, Scuderi C, et al. Cannabidiol in inflammatory
       bowel diseases: an overview. Phytother Res 2013;27(5):633-6.
[65]   De Vries K, Green AJ. Therapeutic use of cannabis. Nurs Times 2012;108(9):12-5.
[66]   Klumpers LE, Beumer TL, van Hasselt JG, Lipplaa A, Karger LB, Kleinloog HD, et al. Novel delta-9-
       tetrahydrocannabinol formulation Namisol has beneficial pharmacokinetics and promising
       phamacodynamics effects. Br J Clin Pharmacol 2011;72(12):810-6.
[67]   Tambaro S, Bortolato M. Cannabinoid-related agents in the treatment of anxiety disorders; current
       knowledge and future perspectives. Recent Pat CNS Drug Discov 2012;7(1):25-40.
                                       Cannabis or marijuana                                              53
[68]   Schier AR, Ribeiro NP, Silva AC, Hallak JE, Crippa JA, Nardi AE, et al. Cannabidiol, a Cannabis sativa
       constituent, as an anxiolytic drug. Rev Bras Psiquiatr 2012;34 Suppl 1:104-10.
[69]   Passie T, Emrich HM, Karst M, Brandt SD, Halpern JH. Mitigation of post-traumatic stress symptoms
       by Cannabis resin: a review of the clinical and neurobiological evidence. Drug Test Anal 2012;4(7-
       8):649-59. doi: 10.1002/dta.1377.
[70]   Stern CA, Gazarini L, Takahashi RN, Guimarães FS, Bertolglio LJ. On disruption of fear memory by
       reconsolidation blockage: Evidence from cannabidiol treatment. Neuropsychopharmacology 2012;
       37(9):2132-42. doi: 10.1038/npp.2012.63.
[71]   Kolliakou A, Ismail K, Atakan Z. Why do psychotic patients use cannabis? Case series. Curr Pharm Des
       18(32):4950-9.
[72]   Grant I, Atkinson JH, Gouaux B, Wilsey B. Medical marijuana: clearing away the smoke. Open Neurol
       J 2012;6:18-25.
[73]   Fattore L, Fratta W. Beyond THC: the new generation of cannabinoid designer drugs. Front Behav
       Neurosci 2011;5:60-4.
[74]   Berkovitz R, Arieli M, Marom E. Synthetic cannabinoids—the new “legal high” drugs. Harefuah
       2011;150(12):884-7.
[75]   Hilderbrand RL. High-performance sport, marijuana, and cannabimimetics. J Anal Toxicol
       2011;35(9):624-37.
[76]   Castellanos D, Thornton G. Synthetic cannabinoid use: recognition and management. J Psychiatr Pract
       2012;18(2):86-93.
[77]   Seely KA, Lapoint J, Moran JH, Fattore L. Spice drugs are more than harmless herbal blends: A review
       of the pharmacology and toxicology of synthetic cannabinoids. Prog Neuropsychopharmacol Biol
       Psychiatry 201239(2):234-43.
[78]   Bebarta VS, Ramirez S, Varney SM. Spice: a new “legal” herbal mixture abused by young active duty
       military personnel. Subst Abus 2012;33(2):191-4.
[79]   Karila Petit A, Cottencin O, Coscas S, Reynaud M. Synthetic drugs: the new low-cost landscape of
       drugs. Rev Prat 2012;62(5):664-6.
[80]   Rosenbaum CD, Carreiro SP, Babu KM. Here today, gone tomorrow….back again? A review of herbal
       marijuana alternatives (K2, Spice), synthetic cathinones (bath salts), Kratom, Salvia divinorum,
       methoxetamie, and piperazines. J Med Toxicol 20128(1):15-32.
[81]   Funada M. Pharmacological properties and dependence liabilities of synthetic cannabinoids. Nihon
       Arukoru Yakubutsu Igakkai Zasshi 2010;45(3):167-74.
[82]   Johnson LA, Johnson RL, Alfonzo C. Spice: a legal marijuana equivalent. Mil Med 2011;176 (6):718-
       20.
[83]   Schneir AB, Cullen J, Ly BT. “Spice” girls: synthetic cannabinoid intoxication. J Emerg Med
       2011;40(3):296-9.
[84]   Ginsburg BC, McMahon LR, Sanchez JJ, Javors MA. Purity of synthetic cannabinoids sold online for
       recreational use. J Anal Toxicol 2012;36(1):66-8.
[85]   De Backer B, Maebe Verstraete AG, Charlier C. Evolution of the content of THC and other major
       cannabinoids in drug-type cannabis cuttings and seedlings during growth of plants. J Forensic Sci
       2012;57(4):918-22. doi: 10.1111/j.1556-4029.2012.02068.
[86]   Köhnemann S, Nedele J, Schwotzer D, Morzfeld J, Pfeiffer H. The validation of a 15 STR multiplex
       PCR for Cannabis species. Int J Legal Med 2012126(4):601-6.
[87]   Mura P, Saussereau E, Brunet B, Goulié JP. Workplace testing of drugs of abuse and psychotropic drugs.
       Ann Pharm Fr 2012;70(3):120-32.
[88]   Salomone A, Gerace E, E’Urso F, Di Corcia D, Vincenti M. Simultaneous analysis of several synthetic
       cannabinoids, THC, CBD, and CBN, in hair by ultra-high performance liquid chromatography tandem
       mass spectrometry. Method validation and application to real samples. J Mass Spectrom 2012;
       47(5):604-12.
[89]   Adverse effects of cannabis. Prescrire Int 2011;20(112):18-23.
[90]   Kalant H. Adverse effects of cannabis on health: an update of the literature since 1996. Prog
       Neuropsychopharmacol Biol Psychiatry 2004;28(5):849-63.
54                                Donald E Greydanus and Joav Merrick
[91]    Greydanus DE, Kaplan G, Baxter LE Sr, Patel DR, Feucht CL. Cannabis: The never-ending, nefarious
        nepenthe of the 21st century: What should the clinician know? Dis Mon 2015;61(4):118-75.
[92]    Lee MH, Hancox RJ. Effects of smoking cannabis on lung function. Expert Rev Respir Med
        2011;5(4):537-46.
[93]    Howden ML, Naughton MT. Pulmonary effects of marijuana inhalation. Expert Rev Respir Med
        2011;5(1):87-92.
[94]    Zarfin Y, Yefet E, Abozaid S, Nasser WM, Finkelstein Y. Infant with altered consciousness after
        cannabis passive inhalation. Child Abuse Negl 2012;36(2):81-3.
[95]    Molly C, Mory O, Basset T, Patural H. [Acute cannabis poisoning in a 10-month-old infant]. Arch
        Pediatr 2012;19(7):729-32. [French]
[96]    Mallaret M, Dal’Bo-Rohrer D, Demattéis M. Adverse effects of marijuana. Rev Prat 2005;55(1):41-9.
[97]    Davis GP, Gunderson EW. Evaluation of chronic cough should consider cannabis use. Am Fam
        Physician 2012;85(7):680-2.
[98]    Benich JJ 3rd, Carek PJ. Evaluation of the patient with chronic cough. Am Fam Physician
        2011;84(8):887-92.
[99]    Bonnet U. Chronic cannabis abuse, delta-9-tetrahydrocannabinol and thyroid function and thyroid
        function. Pharmacopsychiatry 201246(1):35-6.
[100]   Fatma H, Mouna B, Leila M, Radhouane D, Taoufik N. Cannabis: a rare case of acute pancreatitis. Clin
        Res Hepatol Gastroenterol 2013;37(1):e24-5.
[101]   Spadari M, Canioni D, Gregoire E, Drouet G, Bourdon JA, Arditti J, et al. Cannabis body packing: two
        case reports. Clin Toxicol (Phila) 2011;49(9):862-4.
[102]   Simonetto DA, Oxentenko AS, Herman ML, Szostek JH. Cannabinoid hyperemesis: a case series of 98
        patients. Mayo Clin Proc 2012;87(2):114-9.
[103]   Fleig S, Brunkhorst R. Hyperemesis and a high water bill. Z Gastroenterol 2011;49(11):1479-81.
[104]   Galli JA, Sawaya RA, Friedenberg FK. Cannabinoid hyperemesis syndrome. Curr Drug Abuse Rev
        2011;4(4):241-9.
[105]   Wallace EA, Andrews SE, Garmany CL, Jelley MJ. Cannabinoid hyperemesis syndrome: literature
        review and proposed diagnosis and treatment algorithm. South Med J 2011;104(9):659-64.
[106]   Nicolson SE, Denysenko L, Mulcare JL, Vito JP, Chabon B. Cannabinoid hyperemesis syndrome: a
        case series and review of previous reports. Psychosomatics 2012;53(3):212-9.
[107]   Luther V, Yap L. A hot bath to calm what ails you: the Cannabis Hyperemesis Syndrome. Acute Med
        2012;11(1):23-4.
[108]   Wild K, Wilson H. Cannabinoid hyperemesis. Emerg Med J 2012;29(1):67-9.
[109]   Cho CM, Hirsch R, Johnstone S. General and oral health implications of cannabis use. Aust Dent J
        2005;50(2):70-4.
[110]   Maloney WJ. Significance of cannabis use to dental practice. J Mich Dent Assoc 2011;93(11):44-8.
[111]   Maloney WJ. Significance of cannabis use to dental practice. Todays FDA 2012;24(1):43-5.
[112]   Lopes CF, de Angelis BB, Prudente HM, de Souza BV, Cardoso SV, de Azambuja Ribeiro RI.
        Concomitant consumption of marijuana, alcohol, and tobacco in oral squamous cell carcinoma
        development and progression: Recent advances and challenges. Arch Oral Biol 2012;57(8):1026-33.
[113]   Pletcher MJ, Vittinghoff E, Kalhan R, Richman J, Safford M, Sidney S, et al. Association between
        marijuana exposure and pulmonary function over 20 years. JAMA 2012;307(2):173-81.
[114]   Ribeiro A, Ferraz-de-Paula V, Pinheiro ML, Vitoretti LB, Mariano-Souza DP, Quinteiro-Filho WM, et
        al. Cannabidiol, a non-psychotropic plant-derived cannabinoid, decreases inflammation in a murine
        model of acute lung injury: role for the adenosine A (2A) receptor. Eur J Pharmacol 2012;678(1-3):78-
        85.
[115]   Tashkin DP. Airway effects of marijuana, cocaine, and other inhaled illicit agents. Curr Opin Pulm Med
        2001;7(2):43-61.
[116]   Pfeifer AK, Lange P. Pulmonary consequences of marijuana smoking. Ugeskr Laeger
        2006;168(1):1743-6. [Danish].
[117]   Szyper-Kravitz M, Lang R, Manor Y, Lahav M. Early invasive pulmonary aspergillosis in a leukemia
        patient linked to aspergillus contaminated marijuana smoking. Leuk Lymphoma 2001;42(6):1433-7.
                                       Cannabis or marijuana                                             55
[142] Campos DR, Yonamine M, de Morases Moreau RL. Marijuana as doping in sports. Sports Med
      2003;33(6):395-9.
[143] Saugy M, Avois L, Saudan C, Robinson N, Giroud C, Mangin P, et al. Cannabis and sports. Br J Sports
      Med 2006;40 Suppl 1:13-5.
[144] Pope HG Jr., Gruber AJ, Yurgelun-Todd D. The residual neuropsychological effects of cannabis: the
      current status of research. Drug Alcohol Depend 1995;38(1):25-34.
[145] Martin-Santos F, Fagundo AB, Crippa JA, Atakan Z, Bhattacharyya S, Allen P, et al. Neuroimaging in
      cannabis use: a systemic review of the literature. Psychol Med 2010;40(3):383-98.
[146] Crippa JA, Lacerda AL, Amaro E, Busatto Filho G, Zuardi AW, Bressan RA. Brain effects of
      cannabis—neuroimaging findings. Rev Bras Psiquiatr 2005;27(1):70-8.
[147] Schreiner AA, Dunn ME. Residual effects of cannabis use on neurocognitive performance after
      prolonged abstinence: a meta-analysis. Exp Clin Psychopharmacol 2012 Jun 25.
[148] Montgomery C, Seddon AL, Fisk JE, Murphy PN, Jansari A. Cannabis-related deficits in real-world
      memory. Hum Psychopharmacol 2012;27(2):217-25.
[149] Marmorstein NR, Iacono WG, McGue M. Associations between substance use disorders and major
      depression in parents and late adolescent-emerging adult offspring: An adoption study. Addiction
      2012107(11):1965-73. doi: 10.1111/j.1360-0443.2012.03934.x.
[150] Harvey MA, Sellman JD, Porter RJ, Frampton CM. The relationship between non-acute adolescent
      cannabis use and cognition. Drug Alcohol Res 2007;26(3):309-19.
[151] Sundram S. Cannabis and neurodevelopment: implications for psychiatric disorders. Hum
      Psychopharmacol 2006;21(4):245-54.
[152] Schneider M. Puberty as a highly vulnerable developmental period for the consequences of cannabis
      exposure. Addict Biol 2008;13(2):253-63.
[153] Dinieri JA, Hurd YL. Rat models of prenatal and adolescent cannabis exposure. Methods Mol Biol
      2012;829:231-42.
[154] Degenhardt L, Coffey C, Romaniuk H, Swift W, Carlin JB, Hall WD, et al. The persistence of the
      association between adolescent cannabis use and common mental disorders into young adulthood.
      Addiction 2013;108(1):124-33. doi: 10.1111/j.1360-0443.2012.04015.x.
[155] Urban NB, Slifstein M, Thompson JL, Xu X, Grigis RR, Raheja S, et al. Dopamine release in chronic
      cannabis uers: A [(11) C] raclopride positron emission tomography study. Biol Psychiatry 2012;71
      (8):677-83.
[156] Zalesky A, Solowij N, Yücel M, Lubman DI, Takagi M, Harding IH, et al. Effect of long-term cannabis
      use on axonal fibre connectivity. Brain 2012;135(Pt7):2245-55.
[157] Leach LS, Butterworth P. The effect of early onset common mental disorders on educational attainment
      in Australia. Psychiatry Res 2012;199(1):51-7.
[158] Yücel M, Solowij N, Respondek C, Whittle S, Fornito A, Pantelis C, et al. Regional brain abnormalities
      associated with long-term heavy cannabis use. Arch Gen Psychiatry 2008;65(6):694-701.
[159] Abushi H, Akirav I. Short-and long-term cognitive effects of chronic cannabinoids administration in
      late-adolescence rats. PLoS One 2012;7(2):e31731.
[160] Gordon SM, Tulak F, Troncale J. Prevalence and characteristics of adolescent patients with co-occurring
      ADHD and substance dependence. J Addict Dis 2004;23:31-40.
[161] Dennis M, Godley SH, Diamond G, Tims FM, Babor T, Donaldson J, et al. The Cannais Youth
      Treatment (CYT) Study: main findings from two randomized trials. J Subst Abuse Treat 2004;27
      (3):197-213.
[162] Han S, Yang BZ, Kranzler HR, Oslin D, Anton R, Farrer LA, et al. Linkage analysis followed by
      association shown NRG1 associated with cannabis dependence in African Americans. Biol Psychiatry
      2012;72(8):637-44.
[163] Hürlimann F, Kupferschmid S, Simon AE. Cannabis-induced depersonalization disorder in adolescence.
      Cannabis-induced depersonalization disorder in adolescents. Neuropsychobiology 2012;65(3):141-6.
[164] Horwood LJ, Fergusson DM, Coffey C, Patton GC, Tait R, Smart D, et al. Cannabis and depression: an
      integrative data analysis of four Australasian cohorts. Drug Alcohol Depend 2012;126(3):369-78.
[165] Van Dam NT, Bedi G, Earleywine M. Characteristics of clinically anxious versus non- anxious regular,
      heavy marijuana users. Addict Behav 201237(11):1217-23.
                                       Cannabis or marijuana                                             57
[166] Budney AJ. Should cannabis withdrawal disorder be included in DSM-5? Psychiatric Times
      2011;28(2):48-50.
[167] Budney AJ, Hughes JR, Moore BA, Vandrey R. Review of the validity and significance of cannabis
      withdrawal syndrome. Am J Psychiatry 2004;161:1967-77.
[168] Budney AJ, Hughes JR. The cannabis withdrawal syndrome. Curr Opin Psychiatry 2006;19:233-8.
[169] Crowley TJ, MacDonald MJ, Whitmore EA. Cannabis dependence, withdrawal, and reinforcing effects
      among adolescents with conduct symptoms and substance use disorders. Drug Alcohol Depend 1998;
      50:27-37.
[170] Vandrey RG, Budney AJ, Hughes JR, Liguoria A. A within-subject comparison of withdrawal
      symptoms during abstinence from cannabis, tobacco, and both substances. Drug Alcohol Depend
      2008;92(1-3):48-54.
[171] Arendt M, Rosenberg R, Fjordback L, Brandholdt J, Foldager L, Sher L, et al. Testing the self-
      medication hypothesis of depression and aggression in cannabis-dependent subjects. Psychol Med
      2007;37(7):935-45.
[172] Calles JL Jr, Nazeer A. Aggressive and violent behavior. In: Greydanus DE, Calles JL Jr, Patel DR,
      Nazeer A, Merrick J, eds. Clinical aspects of psychopharmacology in childhood and adolescence. New
      York: Nova Science, 2011:106.
[173] Schacht JP, Hutchinson KE, Filbey FM. Associations between cannabinoid-receptor-1 (CNR1)
      variation and hippocampus and amygdale volumes in heavy cannabis users. Neuropsychopharmacology
      201237(11):2368-76. doi: 10.1038/npp.2012.92.
[174] Haney M, Hart CL, Vosburg KD. Marijuana withdrawal in humans: effects of oral THC or divalproex.
      Neuropsychopharmacology 2004;29:158-70.
[175] Milin R, Manion I, Dare G, Walker S. Prospective assessment of cannabis withdrawal in adolescents
      with cannabis dependence: A pilot study. J Am Acad Child Adolesc Psychiatry 2008;47:174-8.
[176] Cornelius JR, Chung T, Martin C, et al. Cannabis withdrawal is common among treatment seeking
      adolescents with cannabis dependence and major depression, and is associated with rapid relapse to
      dependence. Addict Behav 2008;103:787-99.
[177] Gorelick DA, Levin KH, Copersino ML, Heishman SJ, Liu F, Boggs DL, et al. Diagnostic criteria for
      cannabis withdrawal syndrome. Drug Alcohol Depend 2012;123(1-3):141-7.
[178] Gardner EL. Addictive potential of cannabinoids: the underlying neurobiology. Chem Phys Lipids
      2005;31;121(1-2):267-90.
[179] Cooper ZD, Haney M. Cannabis reinforcement and dependence: role of cannabinoid CB1 receptor.
      Addict Biol 2008;13(2):188-95.
[180] Cooper ZD, Haney M. Actions of delta-9-tetrahydrocannabinol in cannabis: relation to use, abuse,
      dependence. Int Rev Psychiatry 2009;2192:104-12.
[181] Gardner EL. Endocannabinoid signaling system and brain reward: emphasis on dopamine. Pharmacol
      Biochem Behav 2005;81(2):263-84.
[182] Gardner EL. Addiction and brain reward and antireward pathways. Adv Psychosom Med 2011;30:22-
      60.
[183] Bossong MG, van Berckel BN, Boellaard R, Zuurman L, Schuit RC, Windhorst AD, et al. Delta 9-
      tetrahydrocannabinol induces dopamine release in the human striatum. Neuropsychopharmacology
      2009;34(3):759-66.
[184] Horey JT, Mariani JJ, Cheng WY, Bisaqa A, Sullivan M, Nunes E, et al. Comparison of substance use
      milestones in cannabis-and cocaine-dependent patients. J Addict Dis 2012;31(1):60-6.
[185] Goldstein RZ, Volkow ND. Dysfunction of the prefrontal cortex in addiction: neuroimaging findings
      and clinical implications. Nat Rev Neurosci 2011;12(11):652-69.
[186] Goldstein RZ, Volkow ND. Drug addiction and its underlying neurobiological basis: neuroimaging
      evidence for the involvement of the frontal cortex. Am J Psychiatry 2002;159(10):1642-52.
[187] Feil J, Sheppard D, Fitzgerald PB, Yücel M, Lubman DI, Bradshaw JL. Addiction, compulsive drug
      seeking, and the role of frontostriatal mechanisms in regulating inhibitory control. Neurosci Biobehav
      Rev 2010;35(2):248-75.
58                               Donald E Greydanus and Joav Merrick
[188] Rotter A, Bayerlein K, Hansbauer M, Weiland J, Sperling W, Kornhuber, et al. Orexin A expression
      and promoter methylation in patients with cannabis dependence in comparison to nicotine-dependent
      cigarette smokers and nonsmokers. Neuropsychobiology 2012;66(2):126-33.
[189] Schafer G, Feilding A, Morgan CG, Agathangelou M, Freeman TP, Valerie Curran H. Investigating the
      interaction between schizotypy, divergent thinking, and cannabis use. Conscious Cogn 2002;21(1):292-
      8.
[190] Galvez-Buccollini JA, Proal AC, Tomaselli V, Trachtenberg M, Coconcea C, Chun J, et al. Association
      between age at onset of psychosis and age at onset of cannabis use in non-affective psychosis. Schizophr
      Res 2012;139(1-3):157-60.
[191] Giovanni M, Giuseppe DI, Gianna S, Domenico DB, Luisa DR, Massimo DG. Cannabis use and
      psychosis: theme introduction. Curr Pharm Des 201218(32):4991-8.
[192] Leweke FM, Koethe D. Cannabis and psychiatric disorders: it is not only addiction. Addict Biol
      2008;13(2):264-75.
[193] Nazeer A, Calles Jr JL. Schizophrenia in children and adolescents. In: Greydanus DE, Calles Jr JL, Patel
      DR, Nazeer A, Merrick J, eds. Clinical Aspects of Psychopharmacology in Childhood and Adolescence.
      New York: Nova Science, 2011:152.
[194] Van Dijk D, Koeter MW, Hijman R, Kahn RS, van den Bring W. Effect of cannabis use on the course
      of schizophrenia in male patients: a prospective cohort study. Schizophr Res 2012;137(1-3):50-7.
[195] Bossong MG, Niesink RJ. Adolescent brain maturation, the endogenous cannabinoid system, and the
      neurobiology of cannabis-induced schizophrenia. Prog Neurobiol 2010;92(3):370-85.
[196] Rapp C, Bugra H, Riecher-Rössler A, Borgwardt S. Effects of cannabis use on human brain structure in
      psychosis: a systematic review combining in vivo structural neuroimaging and post-mortem studies.
      Curr Pharm Des 201218(32):5070-80.
[197] Khan MK, Usmani MA, Hanif SA. A case of self amputation of penis by cannabis induced psychosis. J
      Forensic Leg Med 2012;19(6):355-7.
[198] Barrowclough C, Emsley R, Eisner E, Beardmore R, Wykes T. Does change in cannabis use in
      established psychosis affect clinical outcome? Schizophr Bull 2013;39(2):339-48.
[199] Leweke FM. Anandamide dysfunction in prodromal and established psychosis. Curr Pharm Des
      2012;18(32):5188-93.
[200] Decoster J, van Os J, Myin-Germeys I, De Hert M, van Winkel R. Genetic variation underlying
      psychosis-inducing effects of cannabis: critical review and future directions. Curr Pharm Des
      2012;18(32):5015-23.
[201] Serafini G, Pompili M, Innamorati M, Rihmer Z, Sher L, Girardi P. Can cannabis increase the suicide
      risk in psychosis? A critical review. Curr Pharm Des 2012;18(32):5165-87.
[202] Hermann D, Schneider M. Potential protective effects of cannabidol on neuroanatomical alterations in
      cannabis users and psychosis: a critical review. Curr Pharm Des 2012;18(32):4897-905.
[203] Lev-Ran S, Aviram A, Braw Y, Nitzan U, Ratzoni G, Fennig S. Clinical correlates of cannabis use
      among adolescent psychiatric inpatients. Eur Psychiatr 2012;27(6):470-5.
[204] Auther AM, McLaughlin D, Carrión RE, Naqachandran P, Correll CU, Cornblatt BA. Prospective study
      of cannabis use in adolescents at clinical high risk for psychosis: impact of conversion to psychosis and
      functional outcome. Psychol Med 2012;30:1-13.
[205] Zuardi AW, Crippa JA, Bhattacharyya S, Atakan Z, Martin-Santos R, et al. A critical review of the
      antipsychotic effects of Cannabidiol: 30 years of translational investigation. Curr Phar Des 2012;18
      (32):5131-40.
[206] Bhattacharyya S, Crippa JA, Allen P, Martin-Santos R, Borgwardt S, Fusar-Poli P, et al. Induction of
      psychosis byδ9-tetrahydrocannabinol reflects modulation of prefrontal and striatal function during
      attentional salience processing. Arch Gen Psychiatry 2012;69(1):27-36.
[207] Blum K, Gardner E, Oscar-Berman M, Gold M. “Liking” and “wanting” linked to reward deficiency
      syndrome (RDS): hypothesizing differential responsivity in brain reward circuitry. Curr Pharm Des
      2012;18(1):113-8.
[208] Bailén AJR, Quesada PS, Valladares MA. Decision making in cannabis users. Adicciones 2012;24
      (2):161-72.
                                        Cannabis or marijuana                                               59
[209] Van Ours JC, Williams J. The effects of cannabis use on physical and mental health. J Health Econ
      2012;31(4):564-77.
[210] Schifano F, Martinotti G, Cunniff A, Reissner V, Scherbaum N, Ghodse H. Impact of an 18- month,
      NHS-based, treatment exposure for heroin dependence: Results from the London Area Treat 2000
      Study. Am J Addict 2012;21(3):268-73.
[211] Porath-Waller AJ, Beasley E, Beirness DJ. A meta-analytic review of school-based prevention for
      cannabis use. Health Educ Behav 2010;37(5):709-23.
[212] Teesson M, Newton NC, Barrett EL. Australian school-based prevention programs for alcohol and other
      drugs: A systemic review. Drug Alcohol Rev 2012;31(6):731-6. doi:10.1111/j.1465-3362.2012.
      00420.x.
[213] De Looze M, Harakeh Z, van Dorsselaer SA, Raaijmakers QA, Vollebergh WH, Bogt TF. Explaining
      educational differences in adolescent substance use and early sexual debut: the role of parents and peers.
      J Adolesc 2012;35(4):1035-44.
[214] Yap MB, Reavley NJ, Jorm AF. Young people’s beliefs about the harmfulness of alcohol, cannabis, and
      tobacco for mental disorders: findings from two Australian national youth surveys. Addiction
      2012;107(4):838-47.
[215] Gillespie NA, Lubke GH, Gardner CO, Neale MC, Keanler KS. Two-part random effects growth
      modeling to identify risks associated with alcohol and cannabis initiation, initial average use, and
      changes in drug consumption in s sample of adult, male twins. Drug Alcohol Depend 2012;123(1-
      3):220-8.
[216] Abdoul H, Le Faou AL, Bouchez J, Touzeau D, Lagrue G. Cannabis cessation Interventions offered to
      young French users: predictors of follow-up. Encaphale 2012; 38(2):141-8.
[217] Scott LA, Roxburgh A, Bruno R, Matthews A. Burns L. The impact of comorbid cannabis and
      methamphetamine use on mental health among regular ecstasy users. Addict Behav 2012;37(9):1058-
      62.
[218] Friedmann PD, Green TC, Taxman FS, Harrington M, Rhodes AG, Katz E, et al. Collaborative
      behavioral management among parolees: drug use, crime & re-arrest in the Step’n Out randomized trial.
      Addiction 2012;107(6):1099-108. doi: 10.1111/j1360-0443.2011.03769.x.
[219] Baker AL, Thornton LK, Hides L, Dunlop A. Treatment of cannabis use among people with psychotic
      disorders: a critical review of randomised control trials. Curr Pharm Des 2012;18(32):4923-37.
[220] Buckner JD, Zvolensky MJ, Schmidt NB. Cannabis-related impairment and social anxiety: the roles of
      gender and cannabis use motives. Addict Behav 2012;37(11):1294-7.
[221] Carroll KM, Nich C, Lapaglia DM, Peters EN, Easton CJ, Petry NM. Combining cognitive behavioral
      therapy and contingency management to enhance their effects in treating cannabis dependence: less can
      be more, more or less. Addiction 2012;107(9):1650-9. doi: 10.1111/j.1360-0443.2012.03877.
[222] Hendriks V, van der Schee E, Blanken P. Matching adolescents with a cannabis use disorder to
      multidimensional family therapy or cognitive behavioral therapy: treatment effect moderators in a
      randomized controlled trial. Drug Alcohol Depend 2012;125(1- 2):119-26.
[223] Baker AL, Hides L, Lubman DI. Treatment of cannabis use among people with psychotic or depressive
      disorders: a systemic review. J Clin Psychiatry 2010;71(3):247-54.
[224] Gates PJ, Norberg MM, Copeland J, Digiusto E. Randomised controlled trial of a noval cannabis use
      intervention delivered by telephone. Addiction 2012;107(12):2149-58. doi: 10.1111/j.1360-0443.
      2012.03953.x.
[225] Lindsey WT, Stewart D, Childress D. Drug interactions between common illicit drugs and prescription
      therapies. Am J Drug Alcohol Abuse 2012;38(4):334-43.
[226] Vandrey R, Haney M. Pharmacotherapy for cannabis dependence: how close are we? CNS Drugs
      2009;23:543-53.
[227] Hjorthøj C, Fohlmann A, Nordentoft M. Treatment of cannabis use disorders in people with
      schizophrenia spectrum disorders-a systemic review. Addict Behav 2009;34(6-7):520-5.
[228] Agrawal A, Verweil KJ, Gillespie NA, Health AC, Lessov-Schlaggar CN, Martin CN, et al. The genetics
      of addiction—a translational perspective. Transl Psychiatry 2012;2:e140.
60                              Donald E Greydanus and Joav Merrick
[229] Crippa JA, Derenusson GN, Chagas MH, Atakan Z, Matin-Santos R, Zuardi AW, et al. Pharmacologic
      interventions in the treatment of the acute effects of cannabis: a systemic review of literature. Harm
      Reduct J 2012;9(1):7.
[230] Crippa JA, Hallak JE, Machado-de-Sousa JP, Queiroz RH, Bergamaschi M, Chagus MH, et al.
      Cannabidol for the treatment of cannabis withdrawal syndrome: A case report. J Clin Pharm Ther
      2013;38(2):162-4. doi.10.1111/jcpt.12018.
[231] Vandrey R, Stitzer ML, Mintzer MZ, Huestis MA, Murray JA, Lee D. The dose effects of short-term
      dronabinol (oral THC) maintenance in daily cannabis users. Drug Alcohol Depend 2013;128(1-2):64-
      70.
[232] Haney M, Hart CL, Vosburg KD. Effects of THC and lofexidine in a human laboratory model of
      marijuana withdrawal and relapse. Psychopharmacology (Berl) 2008;197:157-68.
[233] Bedi G, Cooper ZD, Haney M. Subjective, cognitive and cardiovascular dose-effect profile of nabilone
      and dronabinol in marijuana smokers. Addict Biol 2013;18(5):872-81. doi:10.1111/j.1369-1600.
      2011.00427.x.
[234] Kleinloog D, Liem-Moolenaar M, Jacobs G, Klaassen E, de Kam M, Hijman R, et al. Does olanzapine
      inhibit the psychomimetic effects of {Delta} 9-tetrahydrocannabinol? J Psychopharmacol 2012;26
      (10):1307-16.
[235] Sarnyai Z. Oxytocin and neuroadaptation to cocaine. Prog Brain Res 1998;119:449-66.
[236] Baskerville TA, Douglas AJ. Dopamine and oxytocin interactions underlying behaviors: potential
      contributions to behavioral disorders. CNS Neurosci Ther 2010;16(3):e92-123.
[237] McGregor IS, Bowen MT. Breaking the loop: Oxytocin as a potential treatment for drug addiction.
      Horm Behav 2012;61(3):331-9.
[238] Gray KM, Carpenter MJ, Baker NL, Desantis SM, Kryway E, Hartwell KJ, et al. A double- blind
      randomized controlled trail of N-acetylcysteine in cannabis-dependent adolescents. Am J Psychiatry
      2012;169(8):805-12. doi: 10.1176/appi.ajp.2012.12010055.
[239] Weinstein AM, Gorelicki DA. Pharmacologic treatment of cannabis dependence. Curr Pharm Des
      2011;17(14):1351-8.
[240] Pertwee RG. Cannabis and cannabinoids: pharmacology and rationale for clinical use. Foresch
      Komplementarmed 1999;6(Suppl 3):12-5.
[241] Svrakic DM, Lustman PJ, Mallya A, Lynn TA, Finney R, Svrakic NM. Legalization, decriminalization
      & medicinal use of cannabis: a scientific and public health perspective. Mo Med 2012;109(2):90-8.
[242] Beaulieu P, Rice AS. The pharmacology of cannabinoids derivatives: are there applications to treatment
      of pain? Ann Fr Anesth Rhanim 2002;21(6):493-508.
[243] Lee MC, Ploner M, Wiech K, Bingel U, Wanigasekera V, Brooks J, et al. Amygdala activity contributes
      to the dissociative effect of cannabis on pain perception. Pain 2013; 154(1):124-34.
[244] Richardson GA, Larkby C, Goldschmidt L, Day NL. Adolescent initiation of drug use effects of prenatal
      cocaine exposure. J Am Acad Child Adolesc Psychiatry 2013; 52(1): 37-46.
[245] Mercolini L, Mandrioli R, Sorella V, Somaini L, Giocondi D, Serpelloni G, et al. Dried blood spots:
      liquid chromatography-mass spectrometry analysis of delta-9-tetrahydrocannabinol and its main
      metabolites. J Chromatogr A 2013;1271(1):33-40.
[246] Danovitch I, Gorelick DA. State of the art treatments for cannabis dependence. Psychiatr Clin North
      Am 2012;35(2);309-26.
[247] Forray A, Sofuoglu M. Future pharmacologic treatments for substance use disorders. Br J Clin
      Pharmacol 2014;77(2):382-400. doi.10.1111/j.1365-2125.2012.04474.x.
[248] Greydanus DE, Feucht C, Hawver EK. Substance abuse and adolescence. Int J Child Health Hum Dev
      2012;5(2):149-79.
[249] Beaulieu P, Boulanger A, Desroches J, Clark AJ. Medical cannabis: considerations for the
      anesthesiologist and pain physician. Can J Anaesth 2016 Feb 5. [Epub ahead of print]
[250] Yarnell S. The use of medicinal marijuana for posttraumatic stress disorder: A review of the current
      literature. Prim Care Companion CNS Disord 2015;17(3). doi: 10.4088/PCC.15r01786. eCollection
      2015.
                                      Cannabis or marijuana                                             61
[251] Volz MS, Siegmund B, Häuser W. [Efficacy, tolerability, and safety of cannabinoids in
      gastroenterology: A systematic review]. Schmerz 2016;30(1):37-46. doi: 10.1007/s00482-015-0087-0.
      [German].
[252] Koppel BS, Brust JC, Fife T, Bronstein J, Youssof S, Gronseth G, Gloss D. Systematic review: efficacy
      and safety of medical marijuana in selected neurologic disorders: report of the Guideline Development
      Subcommittee of the American Academy of Neurology. Neurology 2014;82(17):1556-63. doi:
      10.1212/WNL.0000000000000363.
[253] Mücke M, Carter C, Cuhls H, Prüß M, Radbruch L, Häuser W. [Cannabinoids in palliative care:
      Systematic review and meta-analysis of efficacy, tolerability and safety]. Schmerz. 2016;30(1):25-36.
      doi: 10.1007/s00482-015-0085-2. [German].
SECTION TWO: PLANT PHARMACOLOGY
In: Cannabis: Medical Aspects                                               ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                      © 2017 Nova Science Publishers, Inc.
Chapter 4
PHARMACOLOGY OF CANNABIS
                                     Mandakini Sadhir*, MD
     Division of Adolescent Medicine, Department of Pediatrics, University of Kentucky,
                       Lexington, Kentucky, United States of America
       Cannabis has been used for recreational purposes around the world. It is derived from the
       plant cannabis sativa which has various other compounds known as cannabinoids. Most
       common form of cannabis used for recreational purpose is marijuana, which is prepared
       from dried flowering tops and leaves. The primary psychoactive component is delta 9-
       tetrahydrocannabinol (δ-9THC), which exerts its physiological and psychological effects
       through its interaction with CB1 and CB2 receptors. Smoking is the most commonly used
       method with onset of effects within minutes after inhalation. Oral ingestion of cannabis has
       varied absorption with delayed onset but longer duration of action. Urine drug screen is the
       most common method for detecting cannabis use. Other cannnabionoids such as
       cannabidiol have been utilized for medicinal purpose and research is ongoing to fully
       understand its role in treatment of various health conditions. Synthetic cannabis has
       emerged as drug of abuse over recent years and poses greater challenge due to serious
       physiological and psychological effects and inability to be detected in standard screening
       tests. This chapter describes main cannabinoids, their mechanism of action and metabolism
       in humans.
                                            INTRODUCTION
Cannabis has been used for recreational purpose around the world for many decades. Its use
however has increased in recent years among adolescents and young adults. Much of this
increase seems to correlate with low risk perception (1, 2). The primary psychoactive
constituent of cannabis is delta 9-tetrahydrocannabinol (δ-9THC) known to have various
physiological and psychological effects. Chronic use leads to dependence and behavioral
disturbances (3). Multiple studies have explored effects of cannabis on various health
conditions, but its medicinal use is currently limited (3, 4). Debate exits over decriminalization
*
    Correspondence: Mandakini Sadhir, MD, Division of Adolescent Medicine, Department of Pediatrics, University
       of Kentucky, Lexington, KY 40536-0284, United States. E-mail: m.sadhir@uky.edu.
66                                      Mandakini Sadhir
and legalization of cannabis with potential to have varied consequences (5). The cannabinoids
are mainly classifed as phytocannabinoids, endocannbinoids and synthetic cannabinoids. The
chapter describes these main cannabinoids, their mechanism of action and metabolism in
humans.
                                PHYTO-CANNABINOIDS
Cannabis is derived from a female plant Cannabis sativa which contains many compounds
known as phyto-cannabinoids or commonly cannabinoids. The principal cannabinoids are delta
9-tetrahydrocannabinol (δ-9THC), cannabidiol (CBD) and cannabinol (CBN) (4). THC initially
isolated in 1964, is a primary psychoactive agent and has been widely studied (6, 7).Over the
years, many new cannabinoid and non- cannabinoid compounds in the plant have been
discovered (4). The number of cannabinoids identified since 2005 has increased from 70 to
104. Other known compounds in the plant have also increased from 400 to 650 (4). The content
of THC varies in different sources and preparations of cannabis. Its content is highest in the
flowering tops and subsequently declines in the leaves, stem and seeds of the plant (8). Most
common form of cannabis that is used for recreational purpose is Marijuana with THC content
from 0.5 % to 5%. It is prepared from the dried flowering tops and leaves (6-8). Another form
of cannabis is hashish with THC content 2–20% and is derived from dried cannabis resin and
compressed flowers (9). The potency of cannabis products has increase significantly from
approx. 3% to 12-16% or higher (percent THC weight/per dry weight of cannabis) over the past
decades due to sophisticated cultivation and plant breeding techniques. The concentration of
THC can also reach about 80% using butane hash oil (4). In the 1960’s and 1970’s, a cannabis
product for example contained about 10 mg of THC. But, in recent years a joint made of other
subspecies of cannabis may contain about 150 mg of THC. Current generation of cannabis user
may be exposed to higher concentrations of THC as compared to those who used cannabis
decades earlier (8).
     Cannabidiol (CBD) is a non-psychoactive cannabinoid obtained from the plant. Over the
years, it has drawn much attention due to its pharmacological activity and therapeutic use (10).
CBD is found to have neuroprotective, analgesic, sedative, anti-emetic, anti-spasmodic, anti-
anxiety and anti-inflammatory properties. Research is ongoing to understand its role in
treatment of various health conditions (10, 11).
                               MECHANISM OF ACTION
Cannabinoids are known to exert their physiological action through G protein coupled receptors
known as CB1 and CB2. These receptors were initially identified in 1990s and have been
extensively studied (12). Activation of these receptors leads to inhibition of adenylyl cyclase
resulting in decreased production of cAMP and changes in ion channel activity. Through these
receptors, cannabinoids hyperpolarize neurons by closing voltage-dependent calcium channels
and activate potassium channels (13). CB1 receptors are present in both central and peripheral
nervous system. They are mostly present in region of brain associated with memory, cognition,
reward, anxiety, pain perception, movement (12-14). These include cortex, hippocampus,
                                Pharmacology of cannabis                                     67
olfactory area, basal ganglia, cerebellum and spinal cord. Few CB1 receptors are found
in the brainstem and thus administration of cannabinoids is not associated with respiratory
depression unlike opioids (8, 13, 14). CB1 receptor activation has also shown to modulate
neurotransmitters in the brain, including glutamate, γ-amino butyric acid (GABA), opioids,
dopamine, and serotonin, resulting in varied effects (8, 9). Role of serotonin receptor 5-HT2A
in causing some of specific effects of THC such as memory deficits, anxiolytic-like effects, and
social interaction has been reported (15). CB2 receptors are present in the cells of immune
system predominantly in the spleen and macrophages and appear to play role in modulation of
cytokine release and immune cell migration (14).
                                 ENDOCANNABINOIDS
Endocannabinoids are endogenous compounds discovered in the 1990s that interact with
cannabinoid receptors and produce similar effects as cannabis (10). The two main endogenous
compounds are anandamide (from the Sanskrit word ananda, meaning bliss) and 2-
arachidonoylglycerol (2AG) (10). These are derivative of the fatty acid arachidonic acid related
to the prostaglandins. Anandamide is partial agonist for both CB1 and CB2 receptors with CB1
efficacy higher than CB2. It produces similar effects to δ-9THC, but is less potent and has
shorter half-life due to rapid metabolism (9, 16). 2-Arachidonoylglycerol was originally
identified in intestinal tissue and is found at much higher levels than anandamide in the brain.
2-AG is thought to be the main endogenous agonist for both CB1 and CB2 receptors (16).
Endocannabinoids also interact with other G protein coupled receptor and ion channels. Some
of the known ion channel are vanilloid receptor-type 1 (TRPV1) activated by anandamide.
Other receptors are several types of potassium channels, alpha7 nicotinic receptors and 5-HT3
receptors (10, 11). The endocannabinoid system comprised of endocannabinoids and its
receptors is thought to mediate various physiological processes and are implicated in health and
disease (16).
                             ABSORPTION OF CANNABIS
The tetrahydrocannabinol and other cannabinoids have varied absorption and effects depending
on dose, route of administration and vehicle. Further physiological factors such as metabolism
and excretion can influence drug concentrations and its subsequent effects (11). Smoking is
most common and widely used method. After inhalation, THC is rapidly absorbed through
lungs and reaches brain quickly with physiological and psychological effects becoming
apparent within seconds to minutes (10, 17). The effects then reach a plateau that can last 2–4
h before slowly declining. While, the amount of THC absorbed is higher with inhalation, the
bioavailability varies according to the depth of inhalation, puff duration and breath-hold (17).
Absorption of THC is variable when ingested. When taken orally, some of the THC is degraded
by liver due to first pass metabolism and converted to 11-hydroxy δ-9THC, which is also a
psychoactive agent (10, 17). The concentration of 11-hydroxy δ-9THC is thus higher in blood
after ingestion than inhalation and may contribute to some of the psychological effects (17).
With oral ingestion, onset of effect and peak concentration of THC in blood is much delayed
68                                       Mandakini Sadhir
(1-2h) (11). Slow gastrointestinal absorption prolongs the duration of action that may last
several hours (8, 9, 11). Other routes of administration for cannabis such as oro-mucosal, rectal
and transdermal have been explored and studied. These routes of administration increase
bioavailability of THC by avoiding first pass metabolism in the liver. Such forms may be
beneficial for therapeutic uses but further research is needed (11).
                                       DISTRIBUTION
Cannabinoids are highly lipophilic and get distributed in adipose tissue, liver, lung and spleen
(9). Following assimilation in blood, concentration of THC in plasma decreases due to
peripheral redistribution particularly in adipose tissues. Sequestration in adipose tissue
prolongs elimination half- life that can last for several days (8, 11). From the adipose tissue,
THC gets slowly released into the blood stream and other body compartments including the
brain. Cannabinoids also cross the placenta, entering into fetal circulation and secreted in breast
milk (9).
for initial screening while GC-MS is used for confirmation (21, 22). The cut off limit for
cannabis metabolites in immunoassay is 50ng/ml; it is 15 ng/ml for GC-MS. Detection of
metabolites in urine varies depending on frequency and duration of cannabis use. It can be
detected anywhere from 3 days (single use) up to 30 days or longer (long term heavy smoker)
(22).
     Detection of THC and its metabolites is influenced by various other factors in addition to
sensitivity and specificity of assays. These factors include route of administration, amount of
cannabinoids consumed and absorbed, body fat content, rate of metabolism and excretion, time
of specimen collection (9, 20).
     There has been growing interest in utilization of oral fluids as an alternative biological
specimen for detection of drugs in forensic and clinical settings (23). Studies have shown oral
fluids to be simple, non-invasive method of specimen collection with advantages of observed
specimen collection, making adulteration difficult. It has shown to have stronger correlation
with blood than urine concentration of cannabis metabolites and can detect recent exposure
(23). It also offers ease of multiple sample collections and lower biohazard risk of specimen
collection. Currently, research on oral fluids in ongoing and further evaluation is needed before
implementing it as a drug screening test (23).
                              SYNTHETIC CANNABINOIDS
Synthetic cannabinoids are a heterogeneous group of compounds originally synthesized for
research purpose to explore endogenous cannabinoid system for possible therapeutic use (24).
However, these compounds became drugs of abuse and were marketed as “designer drugs”. In
the 2000s, synthetic cannabinoids were sold under brand names such as “spice” and “K2,”
labeled as herbal incense. These became very popular drugs of abuse as they could not be
detected by standard screening tests (25).
     The synthetic cannabinoids act on cannabinoid receptors and have greater affinity to CB1
than CB2 receptors. Both animal and in vitro studies have shown that synthetic cannabinoids
are 2-100 times more potent than THC (25). Synthetic cannabinoids are metabolized in the liver
via conjugation and oxidation pathways and have longer elimination half- life (24, 25). Use of
synthetic cannabinoids has resulted in medical and psychiatric emergencies due to their intense
physiological and psychological effects. Some of the common adverse effects include seizures,
myocardial infarction, acute renal failure, anxiety, agitation, psychosis, suicidal ideation, and
cognitive impairment. Long-term or residual effects are currently unknown (26, 27).
                                        REFERENCES
[1]   Johnston LD, O’Malley PM, Miech RA, Bachman JG, Schulenberg JE. Monitoring the future. National
      survey results on drug use, 1975–2014. 2014 Overview. Key findings on adolescent drug use. Ann
      Arbor, MI: University of Michigan, 2015.
[2]   Pacek LR, Mauro PM, Martins SS. Perceived risk of regular cannabis use in the United States from
      2002 to 2012: Differences by sex, age, and race/ethnicity. Drug Alcohol Depend 2015;149:232-44.
70                                           Mandakini Sadhir
[3]    Ammerman S, Ryan S, Adelman WP, Committee on Substance Abuse, Committe on Adolescence. The
       impact of marijuana policies on youth: clinical, research, and legal update. Pediatrics 2015;135(3):769-
       85.
[4]    Madras BK. Update on cannabis and its medicinal use. Belmont, MA: McLean Hospital, Alcohol Drug
       Abuse Research Program, 2015.
[5]    Haase H, Eyle N, Schrimpf J. The international drug control treaties: How important are they to US
       drug reform? New York: Committee on Drugs and the Law, Special Subcommittee on International
       Drug Law and Policy, New York City Bar Association, 2012.
[6]    Robson P. Cannabis. Arch Dis Child 1997;77(2):164-6.
[7]    Hall W, Solowij N. Adverse effects of cannabis. Lancet 1998;352(9140):1611-6.
[8]    Ashton CH. Pharmacology and effects of cannabis: a brief review. Br J Psychiatry 2001;178:101-6.
[9]    Kumar RN,Chambers WA, Pertwee RG. Pharmacological actions and therapeutic uses of cannabis and
       cannabinoids. Anaesthesia 2001;56(11):1059-68.
[10]   Huestis MA. Human Cannabinoid Pharmacokinetics. Chem Biodivers 2007;4(8):1770-804.
[11]   Huestis MA. Pharmacokinetics and metabolism of the plant cannabinoids, delta-9-
       tetrahydrocannabinol, cannabidiol and cannabinol. Handb Exp Pharmacol 2005;168:657-90.
[12]   Hirst R, Lambert D, Notcutt W. Pharmacology and potential therapeutic uses of cannabis. Br J Anaesth
       1998;81(1):77-84.
[13]   Mackie K. Cannabinoid receptors: where they are and what they do. J Neuroendocrinol 2008;20(Suppl
       1):10-4.
[14]   Pertwee RG. Cannabinoid pharmacology: the first 66 years. Br J Pharmacol 2006;147(Suppl 1):S163-
       71.
[15]   Viñals X, Moreno E, Lanfumey L, Cordomí A, Pastor A, de la Torre R, et al. Cognitive impairment
       induced by delta9-tetrahydrocannabinol occurs through heteromers between cannabinoid CB1 and
       serotonin 5-HT2A Receptors. PLoS Biol 2015;13(7):e1002194.
[16]   Maccarrone M, Bab I, Bíró T, Cabral GA, Dey SK, Di Marzo V, et al. Endocannabinoid signaling at
       the periphery: 50 years after THC. Trends Pharmacol Sci 2015;36(5):277-96.
[17]   Sharma P1, Murthy P, Bharath MM. Chemistry, metabolism, and toxicology of cannabis: clinical
       implications. Iran J Psychiatry 2012;7(4):149-56.
[18]   Jiang R, Yamaori S, Takeda S, Yamamoto I, Watanabe K. Identification of cytochrome P450 enzymes
       responsible for metabolism of cannabidiol by human liver microsomes. Life Sci 2011;89(5-6):165-70.
[19]   Watanabe K, Yamaori S, Funahashi T, Kimura T, Yamamoto I. Cytochrome P450 enzymes involved
       in the metabolism of tetrahydrocannabinols and cannabinol by human hepatic microsomes. Life Sci
       2007;80(15):1415-9.
[20]    Musshoff F, Madea B. Review of biologic matrices (urine, blood, hair) as indicators of recent or
       ongoing cannabis use. Ther Drug Monit 2006;28(2):155-63.
[21]   Chiarotti M, Costamagna L. Analysis of 11-nor-9-carboxy-delta(9)-tetrahydrocannabinol in biological
       samples by gas chromatography tandem mass spectrometry (GC/MS-MS). Forensic Sci Int
       2000;114(1):1-6.
[22]   Moeller KE, Lee KC, Kissack JC. Urine drug screening: practical guide for clinicians. Mayo Clin Proc
       2008;83(1):66-76.
[23]   Lee D, Huestis MA. Current knowledge on cannabinoids in oral fluid. Drug Test Anal 2014;6(1-2):88-
       111.
[24]   Wiley J, Marusich J, Huffman J. Moving around the molecule: relationship between chemical structure
       and in vivo activity of synthetic cannabinoids. Life Sci 2014;97(1):55-63.
[25]   Castaneto MS, Gorelick DA, Desrosiers NA, Hartman RL, Pirard S, Huestis MA. Synthetic
       cannabinoids: epidemiology, pharmacodynamics, and clinical implications. Drug and Alcohol
       Dependence. Drug Alcohol Depend 2014;144:12-41.
[26]    Castellanos D, Gralnik LM. Synthetic cannabinoids 2015: An update for pediatricians in clinical
       practice. World J Clin Pediatr 2016;5(1): 16-24.
[27]   National Institute on Drug Abuse. Synthetic cannabinoids: Drug facts. URL:https:// www.drugabuse.
       gov/publications/drugfacts/synthetic-cannabinoids.
In: Cannabis: Medical Aspects                                           ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                  © 2017 Nova Science Publishers, Inc.
Chapter 5
      The efforts to understand the nature of how the consumption of cannabis affects the human
      body are ongoing, complex, and multifaceted. Documentation on the use of cannabis dates
      back thousands of years; however, it is only now with the recent softening of legal
      restrictions that modern research approaches have been able to initiate an appropriate level
      of detailed investigations. For clinicians, researchers and policy makers, this chapter
      reviews the general structure of cannabinoids, the current understanding of cannabinoids
      on cellular systems, the diference between inhalation and oral consumption on cannabinoid
      bioavailability, the variance among purified cannabinoids versus whole plant extract, and
      the potential activities of another prominent family of secondary metabolites found in
      cannabis, the terpenes.
                                         INTRODUCTION
The efforts to understand the nature of how the consumption of cannabis affects the human
body is an ongoing and complex process. Although documentation of cannabis’ use dates back
thousands of years, it was only in the recent amelioration of legal restrictions that allowed
modern research approaches to initiate an appropriate level of detailed investigations, as with
other plant species. For clinicians, researchers and policy makers, this chapter reviews the
general structure of cannabinoids, the current understanding of cannabinoids within cellular
systems, the differences of inhalation and oral consumption on cannabinoid bioavailability, the
therapeutic efficacy of purified cannabinoids versus whole plant extract, and the potential
activities of another prominent family of secondary metabolites found in cannabis, the terpenes.
*
    Correspondence: Jeremy Friedberg, PhD, MedReleaf Corp, Markham Industrial Park, POBox 3040, Markham,
       Ontario, L3R 6C4, Canada. E-mail: jfriedberg@medreleaf.com.
72                      Istok Nahtigal, Alexia Blake, Andrew Hand et al.
Cannabis sativa produces a wide range of secondary metabolites, with the total number of
identified and reported compounds increasing steadily since Gaoni and Mechoulam first
isolated (-)-trans-delta-9-tetrahydrocannabinol (Δ9-THC) in 1964 (8). In total, 545 different
compounds have been isolated, of which 104 belong to a group of compounds unique to
Cannabis sativa, referred to as cannabinoids (9-17) (see Table 1). However, this number is
considered by many researchers to be dynamic and is a subject of debate, with the number of
cannabinoid-like compounds possibly exceeding 130 (18). Most of these compounds are
typically present only in trace quantities, and the pharmacological value of only a small number
has been researched. The focus of this paper on the pharmacological action of Δ9-THC and
cannabidiol (CBD).
                                      Table 1. Predominant cannabinoids with clinical relevance
     The primary cannabinoid that is responsible for the psychotropic effects of Cannabis sativa
is Δ9-THC (19) (see figure 1). Similar to endogenous post-synaptic released endocannabinoids,
anandamide and 2-arachidonoylglycerol, Δ9-THC interacts with and activates G protein-
coupled CB1 and CB2 cannabinoid receptors (20-22). CB1 receptors are found in a high
concentration in many tissue types throughout the body, including most brain regions and the
peripheral nervous system (23), as well as some non-neuronal tissues such as the liver, stomach,
heart, testes, and fat tissue (24-28). Presynaptic activation of CB1 receptors in neuronal tissue
inhibits release of neurotransmitters such as gamma-Aminobutyric acid and glutamate by
releasing βγ-subunits from the G protein complex, leading to inhibition of voltage-gated
calcium channels and vesicle release (29-30). However, while activation of CB1 receptors
typically inhibits release of neuronal transmitters, in vivo activation of CB1 with Δ9-THC has
been observed to occasionally increase release of acetylcholine, dopamine and glutamate in
various regions of the brain in rats (31-34). It is likely that this is due to selective antagonism
by Δ9-THC of endocannabinoids, as reported by Patel and Hillard (35) when observing anti-
anxiolytic effects of Δ9-THC administration in mice. It is this inhibitory-stimulation modulation
of neurotransmitter release mediated by Δ9-THC that is thought to be responsible for the
psychotropic effects of Cannabis, both depressant and excitatory in nature. Cannabidiol,
however, does not share psychotropic activity with Δ9-THC, instead acting as a CB1 inverse
agonist or even antagonist, thereby attenuating in vivo response to Δ9-THC in multiple model
species (36). Cannabinoid CB2 receptors on the other hand, are more typically located on organs
related to the immune system, and when activated attenuate pro-inflammatory responses such
                         The pharmacological properties of cannabis                                75
as cytokine release and immune cell response (37-38) (see Figure 1). There is evidence that
CBD interacts with CB2 receptors as an inverse agonist, leading to the well-documented
reduction of clinical pro-inflammatory markers such as TNF-α, iNOS and COX-2 expression
(39). In addition to the effects on CB2, CBD has also been reported to interact with additional
immune system related receptors. For example, CBD has been found to potently inhibit uptake
of adenosine at A2A receptors, the mechanism by which adenosine signaling terminates,
thereby enhancing the anti-inflammatory effects of adenosine agonists (40). CB2 receptors are
also found in both brain and peripheral neuronal tissue in a lower concentration relative to CB1
receptors, however their role has yet to be elucidated (41).
 Route of
                         Inhalation                          Oral
 Administration
 % Dose Consumed         ~ 50% (loss due to pyrolysis)       100%
 Trajectory to           Lungs – Bronchi-Bronchiole -        Stomach – Small Intestines – Portal
 Circulation             Alveoli                             Vein - Liver
                                                             Absorption (stomach contents,
                         Intake upon inhalation (puff
 Other Factors                                               metabolic rate, genetic variants in
                         duration, intake volume,
 Affecting Uptake                                            CYP 450 enzyme activity, enzyme
                         holding time)
                                                             regulation by other drugs)
 First-Pass Hepatic                                          First-Pass Hepatic Metabolism by
                         Bypassed
 Metabolism                                                  CYP450 enzymes
 Bioavailability         2 – 56%                             < 20%
 Onset                   Immediate                           30 – 90 minutes
 Time of Peak
                         5 – 10 minutes                      1 – 6 hours
 Plasma
 Duration                2 - 4 hours                         4 – 8 hours
    THC is detectable in blood almost immediately after smoking, with peak plasma
concentrations measurable after 5 – 10 minutes (42, 44-46). Reported peak values vary with
administered dose. For instance, one study reported that inhalation of cigarettes containing
1.75% THC (equivalent to 16 mg THC) and 3.55% THC (34 mg THC) resulted in mean peak
plasma concentrations of 84.3 ng/ml and 162.2 ng/ml, respectively (42). However, the range of
measured peak plasma concentrations for the low dose cigarette was 50-129 ng/ml and 76-267
ng/ml for the high dose cigarette.
76                      Istok Nahtigal, Alexia Blake, Andrew Hand et al.
     Such wide ranges are also found when comparing reported bioavailability values. Some
studies have reported the bioavailability of inhaled THC as 30% (46), 10 – 35%(43), and
18%(47). One study comparing the pharmacokinetics of THC between frequent and occasional
users concluded that the bioavailability was 23 – 27% for frequent users, and 10 – 14% for
occasional users (45). These differences arise from variances in smoking technique, with
factors such as puff duration, intake volume, and holding time determining drug intake (42, 43,
48). Furthermore, up to 30% of THC has been shown to be lost during the pyrolysis process,
with additional loss occurring in the side stream smoke and incomplete absorption in the lungs
(43, 45, 49). As a conservative calculation, the bioavailability of THC after smoking is reported
as 2-56% (42, 48).
     Fewer studies have focused exclusively on the PK activity of CBD. One study reported
that the bioavailability of CBD after inhalation was 31%, while others remark on the similarity
in PK activity between THC and CBD (43, 50). However, it has been reported that CBD may
alter the PK activity of THC, and can mediate some of its adverse effects, such as paranoia and
anxiety (42, 50-53). The exact reason for this modulatory effect is unknown, but current
scientific opinion is that CBD inhibits the activity of cytochrome P450 enzymes, which in turn
effects THC metabolism, particularly after oral administration (42, 48, 51).
     The PK activity of cannabis after oral administration is rather different. Absorption is much
slower and irreproducible, with the onset of action ranging between 30 – 90 minutes. Peak THC
plasma concentrations may be reached as early as 1-2 hours after ingestion or as late as 4 – 6
hours (42, 45). Also, the duration of effects is noticeably longer after oral administration than
after inhalation (48).
     Oral administration is known to diminish the bioavailability of both THC and CBD
compared to inhalation. Several studies have reported that the bioavailability of THC after
ingestion is 4-20% (42), 4-12% (43), 3-14% (50), and 6% (52). Similarly, the oral
bioavailability of CBD has been reported as 13 – 19% (54) and 6% (55).
     The major explanation for this reduction in oral bioavailability is that cannabinoids
undergo extensive first pass hepatic metabolism by CYP 450 genes prior to reaching systemic
circulation (42, 43, 50). Oxidation into 11-OH-THC and other metabolites diminishes the
amount of THC that reaches systemic circulation, thereby reducing oral bioavailability. For the
same reason, plasma levels of 11-OH-THC are significantly higher after oral administration
compared to inhalation (43). With inhalation, first pass hepatic metabolism is avoided since the
cannabinoids enter system circulation via the lungs. Overall, these differences in PK activity
allow patients to customize their treatment based on their therapeutic needs. For example, a
patient in need of instant pain relief may prefer to smoke or vape cannabis. Conversely, a patient
with insomnia may be less interested in instant effects, and instead may prefer to ingest
cannabis and experience its effects throughout the night.
oxygen moieties or branching methyl groups. Terpenes and terpenoids are the primary
constituents of the essential oils of plants and flowers (64). They are a chief constituent of the
Cannabis sativa plant; as of 2011, over 200 terpenoids have been identified in cannabis, with
little being known of how they affect the pharmacological properties (65). The synergistic
relationship between terpenes and cannabinoids can occur through four different mechanisms;
i) multi-target physiological effects, ii) pharmacokinetics, iii) bacterial resistance and iv) side
effect modulation. The synergistic potential of terpenes adds weight to the idea that plants can
be better drugs than singular compound derived from them (65).
      Terpenoids are pharmacologically versatile due to their lipophilic nature, enabling
interaction with cell membranes, neuronal and muscle ion channels, neurotransmitter receptors,
G-protein coupled receptors, second messenger systems and enzymes (66). These substances
have immensely broad biochemical effects, influencing some of the most critical enzyme
systems, while affecting neurotransmitter levels and other fundamental processes. These effects
are exactly what pharmaceutical drugs are designed to do. One of the most important and
captivating aspects of these novel compounds is that they are pharmacologically active in
extremely minute quantities well below toxic levels. Terpenoids are bioavailable in high
percentages due to their lipophilic properties, permitting passive migration across biological
membranes and enter the blood stream moving onto influencing activities of the brain, heart or
other organs. Some of the most commonly found terpenes in Cannabis sativa are:
        D-limonene: studies using citrus oils in mice and humans showed profound anxiolytic
         and antidepressant effects (67, 68).
        β-Myrcene: anti-inflammatory, analgesic and sedative properties (69).
        α-Pinene: anti-inflammatory, antibacterial and a bronchodilator, as well as being able
         to counteract short-term memory deficits induced by THC intoxication (65, 68).
        D-Linalool: anxiolytic activity (68, 70).
        β-Caryophyllene: is the most common sesquiterpenoid encountered in cannabis.
     While these compounds are the major representatives by mass, it is important to note that
there are significantly more chemical species present in small quantities each with its own and
compounded effects.
     Plant antioxidants are composed of a broad variety of compounds, such as ascorbic acid,
polyphenolic compounds, and terpenoids. Terpenes are the main components of essential oils,
their anti-oxidative capacity contributes to the beneficial properties of fruits and vegetables.
Three main modes of antioxidant action have been detected to date: (i) quenching of singlet
oxygen, (i) hydrogen transfer, and (iii) electron transfer. Several investigations have studied
reactive oxygen species and the antioxidant activity of monoterpenes and diterpenes or essential
oils in vitro (71). Reactive oxygen species (ROS) are created from free radicals generated
during energy metabolism and by environmental deterioration, inadequate nutrition and
exposure to irradiation and stress involved in the pathological development of many human
diseases such as neurodegeneration, cardiovascular deterioration, diabetes and others. The most
promising strategy to avert oxidative damage caused by these reactive species is the use of
antioxidant molecules. Antioxidants play an important role in defending the body against free
radical attack by delaying or inhibiting the oxidation of lipids or other biomolecules,
preventing, or facilitating the repairing, of the damage to cells (72). These compounds can act
                           The pharmacological properties of cannabis                                       79
                                            CONCLUSION
Cannabis is plant rich with diverse compounds that exhibits a range of effects on human
physiology. These effects are primarily attributed to cannabinoids and terpenes, large families
of metabolites that can interact with many cellular and physiological systems in the body.
Although much research still needs to be done, the effects of these metabolites provides an
important tool in managing a range of clinical symptoms. Among cultivars of the plant, varying
levels of these compounds create different physiological effects and depending on how the
plant is administered to patients, can alter the clinical utility.
                                     ACKNOWLEDGMENTS
      We thank Dean Pelkonen for assistance with graphic design for Figure 1.
                                            REFERENCES
[1]    Andre CM, Hausman JF, Guerriero, G. Cannabis sativa: the plant of the thousand and one molecules.
       Front Plant Sci 2016;7:19.
[2]    Appendino G, Gibbons S, Giana A, Pagani A, Grassi G, Stavri M, et al. Antibacterial cannabinoids from
       cannabis sativa: a structure-activity study. J Nat Prod 2008;71:1427-30.
[3]    Brenneisen, R. Chemistry and analysis of phytocannabinoids and other cannabis constituents. Forensic
       Sci Med Marijuana Cannabinoids 2007:17-49.
[4]    Fellermeier M, Zenk MH. Prenylation of olivetolate by a hemp transferase yields cannabigerolic acid,
       the precursor of tetrahydrocannabinol. FEBS Lett 1998;427:283-5.
[5]    Happyana N, Agnolet S, Muntendam R, Van Dam A, Schneider B, Kayser O. Analysis of cannabinoids
       in laser-microdissected trichomes of medicinal cannabis sativa using LCMS and cryogenic NMR.
       Phytochemistry 2013;87:51-9.
[6]    Sirikantaramas S, Taura F, Tanaka Y, Ishikawa Y, Morimoto S, Shoyama Y. Tetrahydrocannabinolic
       acid synthase, the enzyme controlling marijuana psychoactivity, is secreted into the storage cavity of
       the glandular trichomes. Plant Cell Physiol 2005;46:1578-82.
[7]    Russo, EB. Taming THC: potential cannabis synergy and phytocannabinoid- terpenoid entourage
       effects. Br J Pharmacol 2011;163:1344-64.
[8]    Gaoni Y, Mechoulam R. Isolation, structure, and partial synthesis of an active constituent of hashish. J
       Am Chem Soc 1964:86;1646-7.
[9]    Ahmed SA, Ross SA, Slade D, Radwan MM, Khan IA, ElSohly MA. Structure determination and
       absolute configuration of cannabichromanone derivatives from high potency cannabis sativa.
       Tetrahedron Lett 2008;49:6050-3.
80                         Istok Nahtigal, Alexia Blake, Andrew Hand et al.
[10]   Ahmed SA, Ross SA, Slade D, Radwan MM, Zulfiqar F, ElSohly MA. Cannabinoid ester constituents
       from high-potency cannabis sativa. J Nat Prod 2008;71:536-42.
[11]   Appendino G, Giana A, Gibbons S, Maffie M, Gnavi G, Grassi G, et al. A polar cannabinoid from
       Cannabis sativa var. Carma. Nat Prod Commun 2008;3:1977-80.
[12]   ElSohly MA, Gul W. Handbook of Cannabis. Constituents of Cannabis Sativa. Oxford: Oxford
       University Press, 2014.
[13]   ElSohly MA, Slade D. Chemical constituents of marijuana: the complex mixture of natural
       cannabinoids. Life Sci 2005;78:539-48.
[14]   Pagani A, Scala F, Chianese G, Grassi G, Appendino G, Taglialtela-Scafati. Cannabioxepane, a novel
       tetracyclic cannabinoid from hemp, cannabis sativa L. Tetrahedron 2011;67:3369-73.
[15]   Radwan MM, Ross SA, Slade D, Ahmed SA, Zulfiqar F, ElSohly MA. Isolation and characterization of
       new cannabis constituents from a high potency variety. Planta Med 2008;74:267-72.
[16]   Taglialatela-Scafati O, Pagani A, Scala F, De Petrocellis L, Di Marzo V, Grassi G, et al.
       Cannabimovone, a cannabinoid with a rearranged terpenoid skeleton from hemp. European J Org Chem
       2010;2010:2023.
[17]   Zulfiqar F, Ross SA, Slade D, Ahmed SA, Radwan MM, Zulfiqar A, et al. Cannabisol, a novel Δ9-THC
       dimer possessing a unique methylene bridge, isolated from cannabis sativa. Tetrahedron Lett
       2012;53:3560-2.
[18]   De Meijer. The chemical phenotypes (chemotypes) of cannabis. In: Handbook of Cannabis. Oxford:
       Oxford University Press, 2014.
[19]   Pertwee RG. The central neuropharmacology of psychotropic cannabinoids. Pharmac Ther
       1987;36:189-261.
[20]   Pertwee RG. Cannabis and cannabinoids: pharmacology and rationale for clinical use. Pharm Pharmacol
       Comm 1997;3:539-45.
[21]   Pertwee RG. Cannabis and cannabinoids: pharmacology and rationale for clinical use. Forsch
       Komplementarmed 1999;6:12-15.
[22]   Pertwee RG. Pharmacological actions of cannabinoids. Handb Exp Pharmacol 2005;168:1-51.
[23]   Svíženskáa I, Dubovýa P, Šulcováb A. Cannabinoid receptors 1 and 2 (CB1 and CB2), their distribution,
       ligands and functional involvement in nervous system structures - a short review. Pharmacol Biochem
       Behav 2008;90:501-11.
[24]   Cota D, Marsicano G, Tschop M, Grubler Y, Flachskamm C, Schubert M, et al. The endogenous
       cannabinoid system affects energy balance via central orexigenic drive and peripheral lipogenesis. J
       Clin Invest 2003;112:423-31.
[25]   Gerard CM, Mollereau C, Vassart G, Parmentier M. Molecular cloning of a human cannabinoid receptor
       which is also expressed in testis. Biochem J 1991;279:129-34.
[26]   Mukhopadhyay P, Batkai S, Rajesh M, Czifra N, Harvey-White J, Hasko G, et al. Pharmacological
       inhibition of CB1 cannabinoid receptor protects against doxorubicin-induced cardiotoxicity. J Am Coll
       Cardiol 2007;50:528-36.
[27]   Pazos MR, Tolon RM, Benito C, Rodriguez CF, Gorgojo JJ, Manuel N. Cannabinoid CB1 receptors are
       expressed by parietal cells of the human gastric mucosa. J Histochem Cytochem 2008;56:511-6.
[28]   Teixeira-Clerc F, Julien B, Grenard P, Nhieu JTV, Deveaux V, Li L, et al. CB1 cannabinoid receptor
       antagonism: a new strategy for the treatment of liver fibrosis. Nat Med 2006;12:671-6.
[29]   Blackmer T, Larsen EC, Bartleson C, Kowalchyk JA, Yoon E, Preininger AM, et al. G protein
       betagamma directly regulates SNARE protein fusion machinery for secretory granule exocytosis. Nat
       Neurosci 2005;8:421-5.
[30]   Szabo S, Schlicker E. Effects of cannabinoids on neurotransmission. Handb Exp Pharmacol
       2005;168:327-65.
[31]   Pertwee RG, Ross RA. Cannabinoid receptors and their ligands. Prostaglandins Leukot Essent Fatty
       Acids 2002;66:101-21.
[32]   Pistis M, Ferraro L, Pira L, Flore G, Tamganelli S, Gessa GL, et al. Δ9-Tetrahydrocannabinol decreases
       extracellular GABA and increases extracellular glutamate and dopamine levels in the rat prefrontal
       cortex: an in vivo microdialysis study. Brain Res 2002;6:155-8.
                           The pharmacological properties of cannabis                                      81
[33]   Gardner EL. Endocannabinoid signaling system and brain reward: Emphasis on dopamine. Pharmacol
       Biochem Behav 2005;81:263-84.
[34]   Pisanu A, Acquas E, Feno S, Di Chiara G. Modulation of Δ9-THC-induced increase of cortical and
       hippocampal acetylcholine release by μ opioid and D1 dopamine receptors. Neuropharmacology
       2006;50:661-70.
[35]   Patel S, Hillard CJ. Pharmacological evaluation of cannabinoid receptor ligands in a mouse model of
       anxiety: further evidence for an anxiolytic role for endogenous cannabinoid signaling. J Pharmacol Exp
       Ther 2006;318:304-11.
[36]   Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Δ9-
       tetrahydrocannabinol, cannabidiol and Δ9- tetrahydrocannabivarin. Br J Pharmacol 2008;153:199-215.
[37]   Cabral GA, Staab A. Effects on the immune system. Handb Exp Pharmacol 2005;168:385-423.
[38]   Pacher P, Mechoulam R. Is lipid signaling through cannabinoid 2 receptors part of a protective system?.
       Prog Lipid Res 2011;50:193-211.
[39]   Castillo A, Tolón MR, Fernández-Ruiz J, Romero J, Martinez-Orgado J. The neuroprotective effect of
       cannabidiol in an in vitro model of newborn hypoxic-ischemic brain damage in mice is mediated by
       CB2 and adenosine receptors. Neurobiol Dis 2010;37:434-40.
[40]   Carrier EJ, Auchampach JA, Hillard CJ. Inhibition of an equilibrative nucleoside transporter by
       cannabidiol: a mechanism of cannabinoid immunosuppression. Proc Natl Acad Sci 2006;103:7895-900.
[41]   Pertwee RG, Howlett AC, Abood ME, Alexander PH, Di Marzo V, Elphick MR, et al. Cannabinoid
       receptors and their ligands: beyond CB1 and CB2. Pharmacol Rev 2010;62:588-631.
[42]   Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers 2007;4:1770-804.
[43]   Grotenhermen, Franjo. Pharmacokinetics and pharmacodynamics of cannabinoids. Anesthesiology
       1997;86:24-33.
[44]   Huestis MA, Henningfield JE, Cone EJ. Blood cannabinoids. I. absorption of THC and formation of 11-
       OH-THC and THCCOOH during and after smoking marijuana. J Anal Toxicol 1992;16:276-82.
[45]   Sharma P, Murthy P, Bharath MMS. Chemistry, metabolism, and toxicology of cannabis: clinical
       implications. Iran J Psychiatry 2012;7:149-56.
[46]   Goulle J, Saussereau E, Lacroix C. Delta-9-tetrahydrocannabinol pharmacokinetics. Ann Pharm
       Françaises 2008;66:232-4.
[47]   Mcpartland JM, Russo EB. Cannabis and cannabis extracts: greater than the sum of their parts? J
       Cannabis Therapeutics 2001;14:103-32.
[48]   Abramovici H. Information for health care professionals: cannabis (marihuana, marijuana) and the
       cannabinoids. Health Canada 2013. URL: http://www.hc-sc.gc.ca/dhp-mps/alt_formats/pdf/marihuana/
       med/infoprof-eng.pdf.
[49]   Orens A, Light M, Rowberry J, Matsen J, Lewandowski B. Marijuana equivalency in portion and dosage
       - an assessment of physical and pharmacokinetic relationships in marijuana production and consumption
       in Denver, CO: Colorado Department of Revenue, 2015.
[50]   Nadulski T, Pragst F, Weinberg G, Roser P, Schnelle M, Fronk EE, et al. Randomized, double-blind,
       placebo-controlled study about the effects of cannabidiol (CBD) on the pharmacokinetics of delta9-
       tetrahydrocannabinol (THC) after oral application of THC verses standardized cannabis extract. Ther
       Drug Monit 2005;27:799-810.
[51]   Zhornitsky S, Potvin S. Cannabidiol in humans—the quest for therapeutic targets. Pharmaceuticals
       2012;5:529-52.
[52]   Karschner EL, Darwin WD, Goodwin RS, Wright S, Huestis MA. Plasma cannabinoid
       pharmacokinetics following controlled oral Δ 9-tetrahydrocannabinol and oromucosal cannabis extract
       administration. Clin Chem 2011;57:66-75.
[53]   Todd SM, Arnold JC. Neural correlates of interactions between cannabidiol and Δ 9 -
       tetrahydrocannabinol in mice : implications for medical cannabis. Br J Pharmacol 2016;173:53-65.
[54]   Mechoulam R, Gallily R. Cannabidiol: an overview of some pharmacological aspects. J Clin Pharmacol
       2002;42:11-9.
[55]   Paudel KS, Hammell DC, Agu RU, Valiveti S, Stinchcomb AL. Cannabidiol bioavailability after nasal
       and transdermal application: effect of permeation enhancers intranasal and transdermal delivery of
       cannabidiol. Drug Dev Ind Pharm. 2010;36:1088-97.
82                         Istok Nahtigal, Alexia Blake, Andrew Hand et al.
Chapter 6
           Despite medical cannabis’ capacity for symptom relief and its legality in many
      regions, it still remains a controversial treatment. Our outpatient palliative care oncology
      clinic has experienced an increase in interest from patients to incorporate medical cannabis
      into their symptom management. The aim of this chapter is to better understand the
      demographics, diagnosis, and symptom profile of patients with advanced cancer accessing
      medical cannabis. Methods: A retrospective chart review was undertaken of patients
      accessing medical cannabis at an academic tertiary Symptom Management Clinic for a
      seven month study period in 2014. Results: Of approximately 340 patients seen in clinic,
      43 patients (12.5%) were identified as having obtained authorization for medical cannabis.
      Patients were predominantly older with 74.4% of patients aged 51-75 years, and 4.7% of
      patients were over 75 years of age. They had a wide variety of cancer diagnosis, of which
      GI maligancies (30%) and lung cancer (20.9%) were the most common. Patients
      experienced a multitude of symptoms with pain being the most common (88.4%), followed
      by fatigue, nausea and lack of appetite. Of note, a significant number (39.5%) reported
      already using medical cannabsi at the time of lisencing. Conclusions: The field of medical
      cannabis use is young and much work still needs to be done to understand its place amongst
      symptom management in general and in palliative patients specifically. This small
      retrospective study describes a cohort of pallaitive cancer patients who accessed medical
      cannabis to manage a variety of symptoms.
*
    Correspondence: Marissa Slaven, MD, Faculty of Health Sciences, McMaster University, 699 Concession St,
      Hamilton, Ontario L9H 1Z8, Canada. E-mail: slavenm@hhsc.ca.
86                       Noah Spencer, Erynn Shaw and Marissa Slaven
                                      INTRODUCTION
     Medical cannabis has been in use for thousands of years. In 2737 BCE, Chinese emperor
Shen-Nung wrote that cannabis could be used to treat constipation, gout, rheumatism, and
absent-mindedness (1). The Ancient Egyptians used cannabis to relieve the pain of hemorrhoids
(2), the Ancient Indians for insomnia (3) and the Ancient Greeks for tapeworms (4).
     Despite medical cannabis’ capacity for symptom relief and its legality in many regions
(including Canada), it still remains a controversial treatment due in part to its psychoactive
components. Consequently, there has been a relatively small amount of research on medical
cannabis users. The limited literature available suggests that modern users are primarily middle-
aged (5-7), male (5-12), and Caucasian (5-9, 13). The most commonly studied modern uses for
medical cannabis are to: treat chronic pain (8, 10, 13-15), improve sleep (10, 13), relieve muscle
spasms, headaches, anxiety, and nausea (10, 13, 15), to relieve depression (7, 10, 13), and to
increase appetite (7, 10, 13, 15).
     In 2001, Canada developed a legal system to regulate the use and distribution of medical
cannabis, and in 2014, shifted away from a homegrown-system to licensed commercial
producers. There are currently 31 licensed producers throughout Canada, which has facilitated
prompt access to safe and reliable sources or medical cannabis (16). Despite the improved
access, there remains a paucity of literature to guide its medical use. The literature available on
medical cannabis use is sparse and limited in its applicability. The majority of studies are small
in sample size, geographically specific, and do not focus on oncology or palliative patients.
     As a tertiary outpatient palliative care oncology clinic, we have experienced an increase in
interest from our patients to incorporate medical cannabis into their symptom management
regime. The aim of this chapter is to better understand the demographics, diagnosis, and
symptom profile of patients with advanced cancer accessing medical cannabis in an out-patient
clinic setting.
                                         OUR STUDY
     This study was a retrospective chart review of patients accessing medical cannabis at a Pain
and Symptom Management Clinic, an academic tertiary center practice in Ontario. The clinic’s
caseload fluctuates between 110 and 130, with a total of 233 new consults seen during the study
period (January 1, 2015 – July 31, 2015). Patients attending the clinic must be 18 or older, have
a cancer diagnosis, and a prognosis of <12 months. Patient records are kept as a combination
of electronic records and paper charts.
     Charts were reviewed for patients having obtained medical cannabis authorization during
their attendance at clinic during the study period. A data extraction tool was created and utilized
to ensure consistency when extracting data from charts. Patient information was de-identified
at completion of collection using numerical identifiers. Data was kept on secure computer with
password protected files. This project was approved by local Research Ethics Board and was a
non-funded study.
                Medical cannabis use in an outpatient palliative care clinic                 87
          Patient characteristics                       n = 43        %
          Age
          31-50                                         9             20.9
          51-75                                         32            74.4
          >75                                           2             4.7
          Gender
          Male                                          27            62.8
          Female                                        16            37.2
          Relationship Status
          Married*                                      25            58.1
          Single                                        8             18.6
          Relationship, not married                     3             7.0
          Not specified                                 7             16.3
          Living Situation
          With spouse/partner                           24            55.8
          Alone                                         3             7.0
          Other**                                       5             11.6
          Not specified                                 11            25.6
          Children at home                              6             14.0
        *Common-law marriages included.
        **Other included: with friends and with extended family.
                                          FINDINGS
     Of approximately 340 patients seen in clinic, 43 patients (12.5%) were identified as having
obtained authorization for medical cannabis over the timeline of the review. 20.9% of patients
were aged 31-50 years, 74.4% of patients were aged 51-75 years, and 4.7% of patients were
over 75 years of age. 62.8% of patients were male and 37.2% of patients were female. 58.1%
of patients were married, 18.6% were single, 7.0% were in an unmarried relationship, and
16.3% of patients’ relationship statuses were unspecified. 55.8% of patients were living with a
spouse or partner, 7.0% were living alone, 11.6% were living either with friends or with
extended family, and 25.6% of patients’ living situations were unspecified. 6 patients were
living with children at home (14.0%) (see Table 1).
     Of the cancer diagnoses represented in this sample, the majority were primary
gastrointestinal malignancies (30.2%), and in particular, colorectal, cholangiocarcinoma,
hepatocellular, and pancreatic malignancies. Many other patients were being treated for a
variety of primary cancers, including lung cancer (20.9%), gynecological cancer (14.0%),
breast cancer (4.7%), and prostate cancer (2.3%). Additional diagnoses represented in this study
and classified together as ‘other’ included melanoma, hematological cancer, genitourinary
cancer, connective tissue cancer, bone cancer, or “unknown origin” (25.6%). 2.3% of patients’
diagnosis were unspecified (see Table 2).
88                       Noah Spencer, Erynn Shaw and Marissa Slaven
     With regards to physical symptoms, the majority of patients (88.4%) reported experiencing
pain. Amongst these, 48.8% reported moderate to severe pain, and 39.5% reported mild pain.
Tiredness was present amongst 46.5% of patients assessed for this symptom; however, nearly
half (48.8%) of charts had not reviewed this symptom specifically. While nausea was also a
prevalent symptom, with 30.2% of patients experiencing this, it is interesting to note that a large
portion (51.5%) reported absence of this symptom. Amongst other physical symptoms present
at the time of authorization were reduction in appetite (37.2%), dyspnea (25.6%), reflux
(16.3%), bowel dysfunction (7.0%), and early satiety (4.7%).
     From a mental health standpoint, 11.6% of patients reported depression and 34.9% reported
anxiety. For both symptoms, a significant number of charts did not report specifically on
presence/absence of symptoms (see Table 3).
     Roughly a quarter of patients were tobacco smokers at the time of their licensing (23.3%),
while 27.9% had smoked in the past and quit, and 23.3% were lifelong non-smokers. 25.6% of
patients’ smoking histories were unspecified. 39.5% of patients were current alcohol consumers
at the time of their licensing, 18.6% had a history of consumption, but were not presently
drinking, 14.0% did not have a history of consumption, and 27.9% of patients’ alcohol
consumption histories were unspecified. 11.6% of patients reported they consumed alcohol
heavily or abused alcohol. No patients were current recreational drug users at the time of their
licensing, 2.3% of patients had used recreational drugs in the past and stopped, and 53.5% of
patients reported no recreational drug use at any time. A large portion (44.2%) of patients’
recreational drug use histories were unspecified (see Table 4).
     A significant number of patients were using cannabis already at the time of their licensing
(39.5%). One patient had used cannabis in the past but had since stopped (2.3%), one patient
had never used cannabis (2.3%), and 55.8% of charts did not specify the patients’ cannabis use
histories (see Table 4).
                 Medical cannabis use in an outpatient palliative care clinic          89
  Patient symptoms                             n = 43                           %
  Pain (presence)
  Present                                      38                               88.4
  Absent                                       3                                7.0
  Not specified                                2                                4.7
  Pain (severity)
  None                                         3                                7.0
  Mild                                         17                               39.5
  Moderate to severe                           21                               48.8
  Not specified                                2                                4.7
  Tiredness
  Present                                      20                               46.5
  Absent                                       2                                4.7
  Not specified                                21                               48.8
  Drowsiness
  Present                                      0                                0.0
  Absent                                       1                                2.3
  Not specified                                42                               97.7
  Nausea
  Present                                      13                               30.2
  Absent                                       22                               51.2
  Not specified                                8                                18.6
  Lack of appetite
  Present                                      16                               37.2
  Absent                                       16                               37.2
  Not specified                                11                               25.6
  SOB/dyspnea
  Present                                      11                               25.6
  Absent                                       16                               37.2
  Not specified                                16                               37.2
  Depression
  Present                                      5                                11.6
  Absent                                       14                               32.6
  Not specified                                24                               55.8
  Anxiety
  Present                                      15                               34.9
  Absent                                       8                                18.6
  Not specified                                20                               46.5
  Other symptoms
  Reflux                                       7                                16.3
  Bowel deregulation*                          3                                7.0
  Early satiety                                2                                4.7
  Other**                                      6                                14.0
  Not specified                                27                               62.8
*Constipation or diarrhea.
**Cough, hiccups, trouble concentrating, edema, thrush.
90                         Noah Spencer, Erynn Shaw and Marissa Slaven
  Substance                                  n = 43                        %
  Smoking
  Current smoker                             10                            23.3
  Past smoker*                               12                            27.9
  Non-smoker                                 10                            23.3
  Not specified                              11                            25.6
  Alcohol consumption
  Current consumption                        17                            39.5
  Past consumption*                          8                             18.6
  None                                       6                             14.0
  Not specified                              12                            27.9
  Heavy use/abuse                            5                             11.6
  Substance/recreational drug use
  Current user                               0                             0.0
  Past user*                                 1                             2.3
  None                                       23                            53.5
  Not specified                              19                            44.2
  Cannabis use
  Current user                               17                            39.5
  Past user*                                 1                             2.3
  None                                       1                             2.3
  Not specified                              24                            55.8
*Have used in the past, but do not currently use substance.
                                           DISCUSSION
     Following the introduction of the "Marihuana for medicinal purposes regulations" in 2014,
medical cannabis treatment became readily accessible to palliative patients. In the busiest
outpatient palliative cancer clinic in Ontario, within a few months of change in policy, the rate
of prescribing was found to increase from less than 1% to 12.5%. We undertook this study to
demographics and symptom profile of palliative oncology patients accessing medical cannabis
at our clinic.
     When reviewing the age of our patients, we found our median age of 58 to be older than
the median age of patients in studies involving non-palliative patient population accessing
cannabis, but younger that the age of palliative patients in general. O’Connell et al. (9) had a
median age of 32 years for long term cannabis users seeking medical cannabis in California,
while Swift et al. (12) found a median age of 45 years. Our patient population, however, was
slightly younger when compared to Kirkova et al. (19), who found a median age of 65 years.
     Our study was similar to existing literature regarding a gender differential amongst medical
cannabis users. Our data found 62.8% of users were male; this is in line with Swift et al. (12)
on the characteristics of 128 Australian medical cannabis patients (63% male) and Ogborne et
al. (11) of 49 Ontario medical cannabis users (63.2%). In large scale studies of general palliative
patient populations, the gender differential is less noticeable. For example, Teunissen et al. (17)
in their systematic review of patients with incurable cancer found that of 2,219 patients
                Medical cannabis use in an outpatient palliative care clinic                    91
reviewed, 53% were male. In Riechelmann et al. (18) study of 255 adult cancer patients
attending Ontario palliative care clinics 54% were male. Finally, Kirkova et al. (19) in their
study of 997 American palliative cancer patients found that 55% were male. It is unclear why
for both palliative patients in our study and non-palliative patients using medical cannabis in
the literature the male cohort is larger than the female cohort.
     From a symptom standpoint, pain was the symptom most prevalent amongst our patients,
with 88.4% reporting at least mild pain and 55.3% rating moderate-severe scores. This
symptom is consistent with much of the existing literature looking at medical cannabis use in
the chronic pain population (9, 11, 14, 15). When compared to the literature specific to
palliative care, pain was prevalent in 84% of in the study population of Kirkova et al. (19), 75%
of Riechelmann et al. (18) and 71% of Teunissen et al. (17).
     Although 46.5% of patients in our study presented with tiredness, fatigue was not common
among participants in studies about the general medical cannabis using population. However,
fatigue was the most common symptom among the palliative cancer patients in studies
conducted by Riechelmann et al. (77%) (18) and Teunissen et al. (74%) (17).
     Both nausea and lack of appetite were reported frequently (30.2% and 37.2% respectively)
amongst our patients. The prevalence of these symptoms are consistent with that previously
found in non-palliative patient populations accessing medical cannabis. Bonn-Miller et al. (13)
in a study of medical cannabis users found 38% used cannabis for appetite stimulation and 25%
for nausea. This is also comparable to the study of Harris et al. (7) of a medical cannabis club
members, in which 33% of participants used cannabis primarily to improve appetite. Amongst
the general palliative patient population nausea and lack of appetite were even more prevalent
with 66% of Riechelmann et al. (18) participants reported lack of appetite and 46% reported
nausea.
     Our patients reported anxiety more frequently (34.9%) compared to depression (11.6%). It
is, however, difficult to draw any meaningful conclusions in this area as the non-response rate
amongst our chart review was high (55.8% and 46.5%, respectively).
     When reviewing the prevalence of alcohol and substance use amongst our patients, the
majority of charts had not recorded specifics around this, leading to high non-response rate for
these variables. Therefore, it is difficult to draw meaningful comparisons in this regard, and
more research would be warranted to better understand the backgrounds of medical cannabis
users receiving palliative care.
     Despite this issue of non-response, one interesting finding was that 39.5% of subjects (and
89.5% of reporting subjects) were already using cannabis at the time of their prescription. This
is a positive sign in that it represents a sizable number of patients who sought to begin using
cannabis grown from sanctioned producers. These producers abide by much stricter health and
safety regulations than their non-approved counterparts.
     In addition to the usual limitations resulting from using data from a retrospective chart
review, the small sample size (n = 43) limits the generalizability and applicability of results.
Furthermore, many of the charts reviewed contained incomplete data, further limiting the
quality of the results. We did not review charts on non-cannabis users; as such, it is difficult to
know how our subjects compared to the general patient population of our clinic. Finally, we
only reviewed charts of oncology patients in the palliative phase of illness, so it is unclear how
generalizable our findings are to non-oncology patients and those in earlier phases of their
illness.
92                          Noah Spencer, Erynn Shaw and Marissa Slaven
                                            CONCLUSION
    In seeking to better understand the demographics and symptom profile of palliative
oncology patients accessing medical cannabis, we found that the large majority of our patient
population were over fifty years old with a predominance of married male patients with GI and
lung malignancies. Additionally, although most had a variety of symptoms almost all (88.4%)
were experiencing pain. A significant portion (39.5%) were already using cannabis at time of
authorization.
    The field of medical cannabis use is young and much work still needs to be done to
understand its place amongst symptom management in general and in palliative patients
specifically. Future studies involving larger sample sizes, direct symptom effect, and more in-
depth sociodemographic information would be helpful to further understand the profile of
patients who derive benefit from this substance.
                                            REFERENCES
[1]    Mack A, Joy J. Can marijuana help? In: Mack A, Joy J. Marijuana as medicine? The science beyond
       the controversy. Washington, DC: Nat Acad Press, 2001:14.
[2]    Pain S. The pharoah’s pharmacists. New Scientist. URL: https://www. newscientist.com/
       article/mg19626341.600-the-pharaohs-pharmacists/
[3]    Touw M. The religious and medicinal uses of cannabis in China, India, and Tibet. J Psychoactive Drugs
       1981;13(1):23-34.
[4]    Butrica JL. The medical use of cannabis among the Greeks and Romans. J Cannabis Therapeutics
       2002;2(2):51-70.
[5]    Reiman A. Medical cannabis patients: patient profiles and health care utilization patterns. J Evidence-
       Based Complem Altern Med 2007;12(1):31-50.
[6]    Reiman A. Cannabis as a substitute for alcohol and other drugs. Harm Reduct J 2009;6:35.
[7]    Harris D, Jones RT, Shank R, Nath R, Fernandez E, Goldstein K, et al. Self-reported marijuana effects
       and characteristics of 100 San Francisco medical marijuana club members. J Addict Dis 2000;19(3):89-
       103.
[8]    Zaller N, Topletz A, Frater S, Yates G, Lally M. Profiles of medicinal cannabis patients attending
       compassion centers in Rhode Island. J Psychoactive Drugs 2015;47(1):18-23.
[9]    O’Connell TJ, Bou-Matar CB. Long term marijuana users seeking medical cannabis in California
       (2001-2007): demographics, social characteristics, patterns of cannabis and other drug use of 4117
       applicants. Harm Reduct J 2007;4:16.
[10]   Nunberg H, Kilmer B, Pacula RL, Burgdorf J. An analysis of applicants presenting to a medical
       marijuana specialty practice in California. J Drug Policy Anal 2011;4(1):1.
[11]   Ogborne AC, Smart RG, Adlaf EM. Self-reported medical use of marijuana: a survey of the general
       population. CMAJ 2000;162:1685-6.
[12]   Swift W, Gates P, Dillon P. Survey of Australians using cannabis for medical purposes. Harm Reduct
       J 2005;2:18.
[13]   Bonn-Miller MO, Boden MT, Bucossi MM, Babson KA. Self-reported cannabis use characteristics,
       patterns and helpfulness among medical cannabis users. Am J Drug Alcohol Abuse 2014;40(1):23-30.
[14]   Ashrafioun L, Bohnert KM, Jannausch M, Ilgen MA. Characteristics of substance use disorder
       treatment patients using medical cannabis for pain. Addict Behav 2015;42:185-8.
[15]   Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, et al. Cannabinoids for
       medical use. A systematic review and meta-analysis. JAMA 2015;313(24):2456-2473.
[16]   Health Canada. Medical use of marijuana. URL: http://www.hc-sc.gc.ca/dhp-mps/marihuana/info/
       index-eng.php
                  Medical cannabis use in an outpatient palliative care clinic                           93
[17]   Teunissen SCCM, Wesker W, Kruitwagen C, de Haes HCJM, Voest EE, de Graeff A. Symptom
       prevalence in patients with incurable cancer: a systematic review. J Pain Sympt Manage 2007;34(1):94-
       104.
[18]   Riechelmann RP, Krzyzanowska MK, O’Carroll A, Zimmermann C. Symptom and medication profiles
       among cancer patients attending a palliative care clinic. Support Care Cancer 2007;15(12):1407-12.
[19]   Kirkova J, Rybicki L, Walsh D, Aktas A. Symptom prevalence in advanced cancer: age, gender, and
       performance status interactions. Am J Hosp Palliat Care 2012;29(2):139-145.
[20]   Barclay JS, Owens JE, Blackhall LJ. Screening for substance abuse risk in cancer patients using the
       opioid risk tool and urine drug screen. Support Care Cancer 2014;22:1883-8.
[21]   Hye Kwon J, Hui D, Chisholm G, Bruera E. Predictors of long-term opioid treatment among patients
       who receive chemoradiation for head and neck cancer. Oncologist 2013;18(6):768-74.
In: Cannabis: Medical Aspects                                           ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                  © 2017 Nova Science Publishers, Inc.
Chapter 7
      The use medical cannabis has been increasing in recent years. This paper provides the
      perspectives of four patients with very different clinical backgrounds and their reported
      experiences using medical cannabis. Not all patients respond the same way to cannabis and
      effective use requires a degree of experimentation as these patients’ perspectives illustrate.
      In these perspectives it is important to note the reported effective management of specific
      symptoms, but also of importance is the reported improvement in general well-being and
      vast improvement in reported quality of life.
                                         INTRODUCTION
As the research into the clinical utility of medical cannabis continues, the significant benefits
of using cannabis in patient care is becoming clear. There are varying degrees of effectiveness
that have been reported on specific symptoms and symptom management as it relates to
particular active components in the plant. But, it has become clear that there is more to the use
of the plant as a whole than just its constituting components. Similarly, many patients are
reporting significant benefits on specific symptoms such as pain or nausea while also reporting
improvements is their ability to cope with their symptoms, translating to improvements in their
general wellbeing. Not intended as detailed case reports, this paper provides the perspectives
of four patients with very different clinical backgrounds and their reported experiences using
medical cannabis. The names and details for each of the patients has been altered to protect
their privacy and maintain confidentiality. Consent was obtained from each patient to report on
the information presented in this article.
    Correspondence: Jeremy Friedberg, PhD, MedReleaf Corp, Markham Industrial Park, POBox 3040, Markham,
       Ontario, L3R 6C4, Canada. E-mail: jfriedberg@medreleaf.com.
96                                     Jeremy Friedberg
                                     CASE STORY 1
Jon is a 32-year-old male, actively working as an independent business owner, and has been
diagnosed with post-traumatic stress syndrome (PTSD). He is currently managing both
symptoms of pain and typical symptoms related to his PTSD diagnosis. Prior to his introduction
to cannabis, Jon’s symptoms were being managed by 8 pharmaceutical medications. Since his
introduction to cannabis Jon reports that all of his symptoms are effectively controlled with
self-regulated doses of up to 7-8 g of cannabis per day. He consumes his cannabis primarily
through vaporizing, but also uses extracted oil – adding it to butter to consume with his meals
– tincture, and occasionally he smokes. As a result of this shift to cannabis, he is no longer
using any pharmaceutical medications. Jon has experimented with several cultivars (strains) of
cannabis but reports that four specific cultivars have been most effective. These varieties
include Luminarium (26-29% THC; 0-0.2% CDB), Elevare (21-24%; 0-0.2% CDB), Sedamen
(20-23% THC; 0% CBD), and Eran Almog (28% THC; 0% CBD). Jon reports that his cannabis
regime has helped with both symptom management and general wellbeing. “… it works very
well, helps take the edge off, and helps regulate symptoms of my PTSD.”
                                     CASE STORY 2
Michael is a retired 73-year-old male diagnosed with a lower lumbar spine injury. As a result
of this injury he is coping with symptoms of burning sensations, intense pain in his feet and
legs, and extreme fatigue. In managing these symptoms, his physician has prescribed him
oxycontin, however, varying dosage regimes have had a marginal effect on pain and produced
a side effect that makes him very drowsy. Under physician guidance, Michael began to
supplement his oxycontin regime with cannabis, specifically the cultivars Avidekel (0-1.3%
THC; 15-18% CBD) and Midnight (7-10% THC, 10-13% CBD), at a dosage of 1g per day.
Michael’s preference is for inhalation, specifically smoking and occasional vaporization.
     As a result of his cannabis use, Michael reported that his pain symptoms are significantly
reduced and when inhaled, the burning sensations are immediately soothed. This effect allowed
him to reduce his Oxycontin intake by 60%. Michael indicated that the oxycontin was the
source of his fatigue and this reduction translated into having more energy. “I’m not sleeping
as much during the day… I fall asleep faster at night. More active, able to dance again.”
     Most notably for Michael was his general improvement in his perceived quality of life.
“We (my wife and I) have our lives back! I am able to go out with my wife, I can dance, I can
spend quality time with her and do the things I have always loved to do. It has changed my
whole life. I no longer have to sleep 16 hours a day anymore.”
                                     CASE STORY 3
Rose is a retired 67-year-old female that has been diagnosed with diffused systemic
scleroderma and pulmonary fibrosis. The primary symptoms of her condition are muscle
and nerve pain, and sleep deprivation. To manage these symptoms she is currently on a
prescribed drug regime that includes myfortic, dexilant, motilium, pentoxifylline, oxycontin,
dimenhydrinate (gravol), and hydroxychloroquine. Under the guidance of her family doctor,
Rose is orally consuming cannabis oil extracts that she adds to her food and drinks. She
                       Four patient perspectives on medical cannabis                           97
primarily prepares her cannabis oil doses from cultivars including Avidekel (0-1.3% THC; 15-
18% CBD) for daytime, Eran Almog (28% THC; 0% CBD), Stellio (22-25% THC; 0% CBD),
Sedamen (20-23% THC; 0% CBD), and Luminarium (26-29% THC; 0% CBD) for nighttime.
For her symptoms, Rose has reported that cannabis is providing exceptional pain management
to the extent that she no longer requires the oxycontin. Her cannabis regime is also effectively
managing her nausea and she no longer needs dimenhydrinate. In terms of her quality of life
and wellbeing, Rose is reporting a significant improvement and attributing it to the cannabis.
“It’s given me a bit of my life back. The pain relief is amazing. I had been on so many different
kinds of medications that were not touching on the pain. Avidekel has really helped with my
daytime wellbeing.”
                                      CASE STORY 4
Cole is 25-year-old male, currently employed. In January 2014 while on vacation, Cole dove
into water from a height of 30 feet and hit his head. He was carried out of the water and couldn’t
walk for several weeks. He has been diagnosed with post-concussion syndrome, spinal
concussion, injury to upper cervical atlas bone, shifted his C1 vertebra, and inflammation in
the brain. As a result of these injuries he is coping with several symptoms including migraine
headaches, severe neck pain, back pain, all of which are causing depression. In coping with
these symptoms his physician had prescribed several pain medications that Cole reported as
having little effect, and actually made him feel worse. When low on traditional options, his
physician suggested trying medical cannabis. Cole began experimenting with CBD cultivars,
specifically Avidekel (0-1.3% THC; 15-18% CBD), but eventually switched to cultivars
containing THC as well. He found that the presence of the THC had a more immediate and
effective reduction of his pain. Cole also reported a general loss of appetite and subsequent
weight loss on traditional pain medications, and that the cannabis use provided the pain
management without affecting his appetite. In his own words, “Normal every day activities that
are extremely easy for everyone, like talking or walking, making a bowel movement, or sexual
intercourse, used to be extremely painful.” In addition to his reported success with pain
management from cannabis, his improvement general wellbeing is also of note. “As soon as I
use it I feel immediate relief… Cannabis has affected my general well-being by helping me live
almost completely normally, which at one point I thought would never be possible.”
                                       CONCLUSION
Medical cannabis is now an established option in a physician’s medical toolkit. Its utility is
derived from the whole-plant mixture of active components and it is the relative proportion of
these components that differentiates cannabis cultivars from one other, their subsequent effects
on symptom management, and how the active components are processed by the patient’s
physiology. However, like all medications, not all patients will respond the same way. Thus,
cannabis requires a degree of experimentation as these patients’ perspectives have illustrated.
It is important to note the reported effective management of specific symptoms, but also of
great importance is the patient’s reported improvement in general well-being and vast
improvement in quality of life.
In: Cannabis: Medical Aspects                                                ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                       © 2017 Nova Science Publishers, Inc.
Chapter 8
                                            INTRODUCTION
Cannabis, more commonly known as marijuana, consists of a group of drugs produced from
plants under the genus Cannabis (1). Cannabis plants contain at least 489 distinct compounds
from 18 different chemical classes and more than 70 different phytocannabinoids (2). The
principal cannabinoids include Δ9-tetrahydrocannbinol (Δ9-THC), cannabinol (CBN), and
cannabidiol (CBD) (3).
     The endocannabinoid system is a ubiquitous lipid signalling system found in all vertebrates
(4). It includes the two main cannabinoid receptors, CB1 and CB2 (4). CB1 receptors are densely
distributed in the brain, whereas CB2 receptors are primarily found in the cells of the immune
and hematopoietic systems (4). The endocannabinoid system is involved in a wide range of
physiological processes in the human body, including immune function, appetite regulation,
pain and inflammation, cardiovascular function, bone development and bone density, synaptic
 Correspondence: Hance Clarke, MD, Staff Anesthesiologist, Director of Clinical Pain Services and Medical Director
     of Pain Research Unit, Toronto General Hospital, 200 Elizabeth Street, Toronto, ON Canada. E-mail:
     Hance.Clarke@uhn.ca.
100                             Bonnie Cheung and Hance Clarke
plasticity and learning, psychiatric disease, psychomotor behaviour, memory, and the
regulation of stress and emotional states (5,6).
    Δ9-THC is a partial agonist at both CB1 and CB2 receptors, while CBN is a product of of
Δ9-THC oxidation and only has 10% of the activity of Δ9-THC (3). CBD lacks any detectable
psychoactivity and does not bind to either CB1 or CB2 receptors at physiologically meaningful
concentrations (7). It has been shown to have antioxidative, anti-inflammatory, and
neuroprotective effects independent of the CB1 and CB2 receptors (3). As a result, it has
therapeutic potential with fewer adverse effects than Δ9-THC.
Many patients when assessed in clinic will refer to the dired bud being consumed by the
lisenced producer’s or dispensary’s listed name (e.g., Alaska or Treasure Island). Unfortunately
when pressed further, patients often have no idea the concentartions of THC or CBD within the
plant product. If we are to truly medicalize this industry, these names should be removed and
the percentage of the medicinal ingerdient should be the primary inforamtion used to identify
the products. Patients would then be able to understand the medicinal value of titrating up or
down the medicinal contents. A standard naming system for different types of cannabis strains
should be established. Further complicating the industry, more than 80% of 75 edible cannabis
products purchased within two large medical cannabis markets in the U.S. were inaccurately
labelled with respect to THC content, which may lead to overdosing or underdosing (17), a
significant public safety issue. Moving away from street names would destigmatize both
patients’ and health care providers’ attitudes towards medical cannabis. Although Canada
legalized cannabis for medical use in 2001, many users still report experiencing stigma from
family, friends, health care providers, and law enforcement, as well as experiencing guilt or
discomfort in using something “illicit” (18). The stigma of medical cannabis acts as a
significant barrier to its acceptability and use. In a qualitative study looking at barriers of access
for Canadian cannabis users, approximately half of the respondents wanted to discuss medical
cannabis with a physician but did not, with the most frequent reason being that they did not feel
comfortable enough to do so (19). Ensuring that the naming and labelling clearly reflects the
contents and indications of the different strains of medical cannabis would likely also improve
communication between the patient and prescriber, such that patients would better understand
the reasons for being prescribed a specific strain of medical cannabis.
2) Standardize dosing
More research regarding the pharmacokinetics as well as into the safe and effective dosing of
medical cannabis should be conducted in an effort to create standard dosing guidelines. Unlike
the majority of prescription medications that consist of single active compounds, cannabis
contains more than 100 cannabinoids, terpenoids, and flavonoids (2). According to Health
Canada, precise dosages for medical cannabis have not yet been established and only rough
dosing guidelines for smoked or vapourized cannabis have been published (9). Dosages for
orally administered products in humans are yet to be established (9). This calls for further
research into how cannabis is metabolized in patients and the factors that determine individual
differences in metabolism.
Currently, there are no clinical guidelines that exist to monitor patients who are taking cannabis
for therapeutic purposes (9). Cannabis continues to be one of the most used illicit drugs;
however, it can lead to physical and psychological dependence (20). Careful consideration must
be taken in certain patient populations, including patients under the age of 25 (21), patients
with severe cardiopulmonary disease (22), smoked cannabis in patients with respiratory
102                                 Bonnie Cheung and Hance Clarke
insufficiency (23), patients with history of psychiatric disorders (24), patients with severe liver
disease (25), and pregnant women (26). While the majority of adverse events from cannabis
are not serious, serious adverse effects of cannabis include impairment in brain connectivity
associated with cannabis in adolescence (27), earlier onset of psychosis (28), and the more
recently discovered cannabinoid hyperemesis syndrome (29). Moreover, there is much less
information on the adverse effects of the medical use of cannabis as compared to the adverse
effects of recreational use (9). This research gap must be addressed if we are to create proper
clinical guidelines for monitoring patients on medical cannabis.
                                           CONCLUSION
In conclusion, standardizing the naming, dosing, and monitoring of medical cannabis will
drastically improve the safety of medical cannabis administration and the stigma surrounding
its use. Currently the public is at the mercy of an industry that lacks stringent regulation. With
the coming legalization of cannabis in Canada, it is imperative that research be conducted with
the patients’ best interests in mind.
                                     ACKNOWLEDGMENTS
Dr. Clarke is supported by a Merit Award from the Department of Anesthesia at the University
of Toronto.
                                           REFERENCES
[1]    Small E, Cronquist A. A practical and natural taxonomy for cannabis. Taxon 1976;25(4):405.
[2]    Elsohly MA, Slade D. Chemical constituents of marijuana: the complex mixture of natural cannabinoids.
       Life Sci 2005;78(5):539–48.
[3]    Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R. Non-psychotropic plant cannabinoids: new
       therapeutic opportunities from an ancient herb. Trends Pharmacol Sci 2009;30(10):515–27.
[4]    Rodríguez de Fonseca F, Del Arco I, Bermudez-Silva FJ, Bilbao A, Cippitelli A, Navarro M. The
       endocannabinoid system: physiology and pharmacology. Alcohol Alcohol Oxf Oxfs 2005;40(1):2–14.
[5]    Serrano A, Parsons LH. Endocannabinoid influence in drug reinforcement, dependence and addiction-
       related behaviors. Pharmacol Ther 2011;132(3):215–41.
[6]    Aggarwal SK. Cannabinergic pain medicine: a concise clinical primer and survey of randomized-
       controlled trial results. Clin J Pain 2013;29(2):162–71.
[7]    Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Δ9‐
       tetrahydrocannabinol, cannabidiol and Δ9‐tetrahydrocannabivarin. Br J Pharmacol 2008;153(2):199–
       215.
[8]    Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, et al. Cannabinoids for
       medical use: a systematic review and meta-analysis. JAMA 2015;313(24):2456.
[9]    Health Canada. Information for health care professionals: cannabis (marihuana, marijuana) and the
       cannabinoids. Health Canada. Accessed 2016 Jul 26. URL: http://www.hc-sc.gc.ca/dhp-mps/alt_
       formats/pdf/marihuana/med/infoprof-eng.pdf.
[10]   Martín-Sánchez E, Furukawa TA, Taylor J, Martin JLR. Systematic review and meta-analysis of
       cannabis treatment for chronic pain. Pain Med 2009;10(8):1353–68.
                               Safety concerning medical cannabis                                       103
[11]   Richards BL, Whittle SL, Buchbinder R. Neuromodulators for pain management in rheumatoid arthritis.
       Cochrane Database Syst Rev 2012;1:CD008921.
[12]   Deshpande A, Mailis-Gagnon A, Zoheiry N, Lakha SF. Efficacy and adverse effects of medical
       marijuana for chronic noncancer pain: Systematic review of randomized controlled trials. Can Fam
       Physician Médecin Fam Can 2015;61(8):e372–81.
[13]   Lakhan SE, Rowland M. Whole plant cannabis extracts in the treatment of spasticity in multiple
       sclerosis: a systematic review. BMC Neurol 2009;9:59.
[14]   Koppel BS, Brust JCM, Fife T, Bronstein J, Youssof S, Gronseth G, et al. Systematic review: efficacy
       and safety of medical marijuana in selected neurologic disorders: Report of the Guideline Development
       Subcommittee of the American Academy of Neurology. Neurology 2014;82(17):1556–63.
[15]   Honourable Jane Philpott. Plenary statement for the Honourable Jane Philpott Minister of Health -
       UNGASS on the World Drug Problem. United Nations General Assembly Special Spession on the
       World Drug Problem. Accessed 2016 Aug 02. URL: http://news.gc.ca/web/article-en.do?nid=1054489.
[16]   Government of Canada. Marihuana for medical purposes regulations Government of
       Canada Department of Justice. Accessed 2016 Aug 02. URL: http://www.laws-lois.justice.gc.ca/eng/
       regulations/SOR-2013-119/FullText.html.
[17]   Vandrey R, Raber JC, Raber ME, Douglass B, Miller C, Bonn-Miller MO. Cannabinoid dose and label
       accuracy in edible medical cannabis products. JAMA 2015;313(24):2491.
[18]   Bottorff JL, Bissell LJ, Balneaves LG, Oliffe JL, Capler NR, Buxton J. Perceptions of cannabis as a
       stigmatized medicine: a qualitative descriptive study. Harm Reduct J 2013;10(1):2.
[19]   Belle-Isle L, Walsh Z, Callaway R, Lucas P, Capler R, Kay R, et al. Barriers to access for Canadians
       who use cannabis for therapeutic purposes. Int J Drug Policy 2014;25(4):691–9.
[20]   Lichtman AH, Martin BR. Cannabinoid tolerance and dependence. Handb Exp Pharmacol
       2005;(168):691–717.
[21]   Health Canada. Consumer information - cannabis (marihuana, marijuana). Government of Canada.
       Accessed 2016 Aug 02. URL: http://www.hc-sc.gc.ca/dhp-mps/alt_formats/pdf/marihuana/info/cons-
       eng.pdf.
[22]   Mathew RJ, Wilson WH, Davis R. Postural syncope after marijuana: a transcranial Doppler study of the
       hemodynamics. Pharmacol Biochem Behav 2003;75(2):309–18.
[23]   Tetrault JM, Crothers K, Moore BA, Mehra R, Concato J, Fiellin DA. Effects of marijuana smoking on
       pulmonary function and respiratory complications: a systematic review. Arch Intern Med 2007;167
       (3):221–8.
[24]   Moore TH, Zammit S, Lingford-Hughes A, Barnes TR, Jones PB, Burke M, et al. Cannabis use and risk
       of psychotic or affective mental health outcomes: a systematic review. Lancet 2007;370(9584):319–28.
[25]   Tarantino G, Citro V, Finelli C. Recreational drugs: a new health hazard for patients with concomitant
       chronic liver diseases. J Gastrointest Liver Dis 2014;23(1):79–84.
[26]   Richardson GA, Ryan C, Willford J, Day NL, Goldschmidt L. Prenatal alcohol and marijuana exposure:
       effects on neuropsychological outcomes at 10 years. Neurotoxicol Teratol 2002;24(3):309–20.
[27]   Zalesky A, Solowij N, Yücel M, Lubman DI, Takagi M, Harding IH, et al. Effect of long-term cannabis
       use on axonal fibre connectivity. Brain J Neurol 2012;135(7):2245–55.
[28]   Large M, Sharma S, Compton MT, Slade T, Nielssen O. Cannabis use and earlier onset of psychosis: a
       systematic meta-analysis. Arch Gen Psychiatry 2011;68(6):555–61.
[29]   Wallace EA, Andrews SE, Garmany CL, Jelley MJ. Cannabinoid hyperemesis syndrome: literature
       review and proposed diagnosis and treatment algorithm. South Med J 2011;104(9):659–64.
In: Cannabis: Medical Aspects                                             ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                    © 2017 Nova Science Publishers, Inc.
Chapter 9
       In recent years cannabis has been used for various medical purposes. The most interesting
       benefit may be related to chemotherapy-induced nausea and vomiting, with cannabis being
       used as an antiemetic in combination with other agents to combat a troublesome side-effect
       of cancer treatment. Medical oncologists are split between whether cannabis should be
       used medicinally because of the quality of evidence. Although patients advocate the benefit
       of cannabis and studies conducted before the 2000s show its efficacy, there have been no
       phase III RCT’s conducted in the last five years that compare cannabis to modern day
       antiemetic regimens. It is critical that phase III trials are conducted to create guidelines for
       future users who will be interested in trying medical cannabis.
                                           INTRODUCTION
Legalization of cannabis has been a hot topic over the past year in Canada with push for
legislation to move forward in the upcoming year. Already in the United States 23 states have
legalized cannabis for medicinal use and four others have completely legalized for recreational
use. Cannabis has been shown to be beneficial in mitigating chronic pain in HIV/AIDS and
cancer patients, and those with fibromyalgia and arthritis. There is evidence that cannabis can
be used to prevent weight loss and increase appetite, minimize seizures for those with epilepsy,
and it can be used to relieve stress and anxiety, especially those with post-traumatic stress
disorder (PTSD). The most interesting benefit may be related to chemotherapy-induced nausea
and vomiting, with cannabis being used as an antiemetic in combination with other agents to
combat a troublesome side-effect of cancer treatment. Medical oncologists are split between
*
    Correspondence: Carlo DeAngelis, PharmD, RPh, Department of Pharmacy, Sunnybrook Health Sciences Centre,
       2075 Bayview Avenue, Toronto, ON, Canada. E-mail: carlo.deangelis@sunnybrook.ca.
106                            Jordan Stinson and Carlo DeAngelis
whether cannabis should be used medicinally because of the quality of evidence mainly
conducted in the 1980s and 1990s, however patients time and again support having access to
medical cannabis and find it very beneficial to manage their disease or condition.
     The issue of legalizing dried cannabis for medicinal use in Canada was kick-started in 2000
and by 2001 the Government of Canada adopted the Marijuana Medical Access Regulations
(MMAR), which recognized medical cannabis as a treatment option for various medical
conditions, including chemotherapy-induced nausea and vomiting (CINV) (1). Applicants
applied directly to Health Canada for authorization to possess, and had the option to grow their
own cannabis or purchase directly from Health Canada (1, 2). As of March 31st, 2014 the
Government of Canada repealed the MMAR and replaced these regulations with the Marijuana
for Medical Purposes Regulations (MMPR), which removed Health Canada from the patient
approval process and eliminated authorization for patients to grow their own cannabis. Patients
now receive their medical cannabis from commercial producers licensed by Health Canada (1,
2). Along with the availability of dried cannabis, two other oral synthetic agents have been
developed and approved for the indication of chemotherapy-induced nausea and vomiting
(CINV); dronabinol (Marinol®), a stereoisomer of delta-9 –THC, and nabilone (Cesamet®), a
synthetic analogue of delta-9-THC. However, in 2012 dronabinol was withdrawn from the
Canadian market for unknown reasons which left nabilone as the only oral agent currently
available. This commentary will explore the currently published data on synthetic and
naturally-derived cannabis to answer the question, is there a role for medical cannabis in the
treatment of chemotherapy-induced nausea and vomiting? We will also shed light on the
opinions of healthcare providers including medical oncologists, pharmacists, and most
importantly the patient.
    Before the year 2000 it appeared that dried cannabis was an effective treatment option for
CINV with manageable side-effects, but what about synthetic oral agents? A systematic review
by Tramer and colleagues published in the BMJ in 2001 looked at 30 randomized trials of
nabilone (16), dronabinol (13), and levonantradol (1) compared to other antiemetics such as
prochlorperazine, metoclopramide, domperidone, and placebo (9). Data from over 1300
108                           Jordan Stinson and Carlo DeAngelis
patients was collected and analyzed. In trials where cannabinoids were compared to placebo,
complete control of nausea was 70% versus 57%, respectively. Complete control of vomiting
was 66% versus 36%, respectively. In trials where cannabinoids were compared to active
controls, complete control of nausea was 59% versus 43%, respectively. Complete control of
vomiting was 57% versus 45%, respectively. Patients preferred cannabinoids over placebo 76%
of the time and against active controls patients still preferred using cannabinoids 61% of the
time. Side-effect profiles occurring in greater than 50% of patients included drowsiness,
sedation and dizziness.
     In 2007 Meiri and colleagues conducted the first study to compare synthetic cannabinoids
with a 5HT3 receptor antagonist for patients receiving moderately to highly emetogenic
chemotherapy (10). In this double-blinded, randomized placebo trial 64 patients received either
ondansetron, dronabinol, or a combination of both drugs. Total response was defined as no
vomiting and less than 5mm on a 0-100mm on the visual analog scale (VAS). Total response
was achieved in 54% of patients that received dronabinol, 58% in patients that received
ondansetron, and 47% in patients that received dronabinol and ondansetron combined. No
statistically or clinically significant difference was observed between both drugs, and total
response for cannabinoid use was less than Tramer and colleagues’ systematic review.
     The conclusion from studies conducted before the new millennium suggested that smoked
cannabis and oral THC capsules were at least comparable and equally effective to antiemetics
such as prochlorperazine and ondansetron, although validity of these studies are questionable
compared to todays’ standards. In 2015 Smith and colleagues conducted a detailed Cochrane
review on the use of cannabinoids for management of chemotherapy-induced nausea and
vomiting (11). In their findings they concluded that cannabis-based medications may be useful
in treating refractory CINV, however the quality of evidence was graded as low and did not
reflect current chemotherapy and antiemetic treatment regimens.
                            ONCOLOGIST PERSPECTIVE
In 1990 an anonymous, random-sample survey was sent out to the members of the American
Society of Clinical Oncology (ASCO) concerning antiemetic use of dried cannabis in cancer
patients. In that survey over 1000 responses were received and a slight majority (54%) thought
cannabis should be available by prescription. As a group, respondents also considered smoked
cannabis to be somewhat more effective than oral synthetic THC drug dronabinol (12). This
survey was the first to demonstrate that oncologists’ experience with medical cannabis was
more favourable then the opinion of the Food and Drug Administration (FDA) at the time.
     In 1997 another survey was mailed to members of ASCO with a similar response rate of
over 1000 medical oncologists (13). This time however recipients were asked whether the DEA
should reschedule dried cannabis to allow it to be prescribed as medicinal cigarettes. Only 28%
of respondents favoured the rescheduling of cannabis as prescription medicine compared to the
54% in 1990 survey. A survey of physicians from multiple disciplines in 2005 found that only
36% were in favour of legalization of medical cannabis (14).
          Medical cannabis in the treatment of chemotherapy-induced nausea…                   109
                             PHARMACIST PERSPECTIVE
Although a pharmacist is not currently involved in the prescribing of medical cannabis in
Canada, new legislation in the upcoming year may change the way patients can access the drug
and more training to educate pharmacists on the use and side-effects will be necessary. Over
the past two decades the amount of research conducted on the use of medical cannabis for
nausea and vomiting, pain, and appetite has resulted in the discovery of multiple routes of
administration including edibles, vaporization, and oils (15). All these routes involve different
pharmacodynamics and pharmacokinetics, but further research on safety and standardization
of strengths is required.
         In the United States the American Society of Health-System Pharmacists (ASHP) is
opposed to the preparation and distribution of medical cannabis but with more states legalizing,
pharmacists are more likely to be asked by patients about drug interactions or disease impact
with regards to their medical cannabis (16). A study conducted by Moeller at al. measured 311
pharmacy students’ knowledge of medical cannabis and their attitude towards it. A moderate
59% of students felt that cannabis should be legalized in all states for medicinal use, and almost
all students did not feel comfortable answering patients’ questions about efficacy, safety, and
drug interactions. 91% of students were aware that medical cannabis was permitted for treating
chemotherapy-related side-effects. The results from this study show that pharmacy students
would benefit from courses on medical cannabis with focus on screening patients, prescribing,
and counseling.
                                PATIENT PERSPECTIVE
Regardless of whether medical cannabis is beneficial for patients suffering from nausea and
vomiting, pain, or lack of appetite the negative stigma of using cannabis still remains. This
stigma comes from the fact that the majority of cannabis use is recreational and not for
medicinal use. A 2004 national survey showed that only 28% of users in Canada reported using
cannabis for medical reasons (17). The negative image of smoking a joint has been reinforced
by film and television labeling users as “potheads”, and public opinion is that cannabis is an
illegal substance (18). However, when ill patients are asked why they use cannabis it paints a
different picture.
          A study by Bottorff and colleagues interviewed 23 individuals with various medical
conditions including HIV/AIDS, fibromyalgia, arthritis, anxiety/mood disorders, cancer,
epilepsy, and chronic pain. To these individuals cannabis was seen as life preserving, an
adjuvant therapy to their disease, and a means of self-management (19). Significant gender
differences were observed with men focusing more on the physical health benefits of smoking
cannabis, while women found improvements in mental health to be the most beneficial. Both
genders shared the idea that cannabis enabled them to take control of their health by choosing
a drug they perceived to be safer and more effective than their prescription medication (19).
          When looking at the patient perspective not only is the psychological aspect of
consuming cannabis important, but also how the patient consumes the product. While the
majority of patients take prescription medication in pill form and ingest orally, there are many
routes of administration for cannabis that make it personalized and completely dependent on
110                             Jordan Stinson and Carlo DeAngelis
the patient to choose. While clinical trials in the 1980s and 1990s involved smoking cannabis
or ingesting THC capsules, patients now have the option to vaporize their cannabis or consume
them in an edible form. With the invention of the vaporizer it is now possible to heat cannabis
to a temperature that vaporizes the cannabinoids while avoiding combustion, which reduces
carbon monoxide and tar generation (15). Peak plasma concentrations of THC are similar for
those that use a vaporizer versus those that smoke cannabis (15). The International Association
for Cannabinoid Medicines (IACM) performed a survey completed by 953 participants world-
wide in over 30 countries on the preference for route of administration. Not surprisingly 92%
of participants tried smoking cannabis with only 72% preferring it as their route of choice.
Vaporizers were the preferred method for 50% of the 450 users who tried it at least once (20).
The participants in this survey who vaporized or smoked cannabis were heavy users consuming
up to 3 grams daily. Top three indications of use were chronic pain (30%), anxiety (18%), and
appetite/weight loss (11%). The most interesting aspect from this survey was that less than 10%
of participants had ever tried dronabinol or nabilone, and for those that did only 23% and 7%
preferred dronabinol and nabilone, respectively (20).
                                  FUTURE DIRECTIONS
     Earlier this year a systematic review of current cannabinoid use for chemotherapy-induced
nausea and vomiting concluded that cannabinoids cannot be recommended as first-line
antiemetics. The authors also stated that due to the lack of randomized controlled trials (RCT)
and safety concerns, medicinal cannabis cannot be recommended for management of CINV
(21). There lies the current problem with medical cannabis. Although patients advocate the
benefit of cannabis and studies conducted before the 2000s show its efficacy, there have been
no phase III RCT’s conducted in the last five years that compare cannabis to modern day
antiemetic regimens. Instead of the Canadian Medical Association (CMA) joining others in
discouraging the prescribing of cannabis, attributing their decision to a lack of clinical evidence
on efficacy, mechanism of action, and potential harm (22, 23) they should be actively
conducting the necessary trials to show the therapeutic benefits of medical cannabis. Physicians
have a responsibility to make clinical decisions in accordance with the best available scientific
evidence and cannot ignore the fact that many drugs used in place of cannabis also have harmful
side-effects that patients must tolerate, with mechanisms of actions that are not fully understood
(24). It is critical that phase III trials are conducted to create guidelines for future users who
will be interested in trying medical cannabis if legislation is passed next year. Future research
should look at the therapeutic benefits of different strains and strengths of dried cannabis to
meet the needs of the user. Ultimately, if cannabis provides physiological or psychological
relief from a medical condition who are we to deny these people access.
                                       REFERENCES
[1]   Canadian Centre on Substance Abuse. Marijuana for medical purposes. Canadian Centre on
      Substance Abuse 2015. URL: http://www.ccsa.ca/Resource%20Library/CCSA-Medical-Purposes-
      Mar ijuana-Policy-Brief-2015-en.pdf
           Medical cannabis in the treatment of chemotherapy-induced nausea…                                111
Chapter 10
       Many patients with advanced cancer experience decreased appetite and involuntary weight
       loss. Weight loss in these patients is rarely recognized, assessed, or actively managed, and
       thus represents an important unmet need in the clinical management of cancer-associated
       anorexia and cachexia. Cannabis has been used as an appetite stimulant since 300 AD and
       presents as a viable therapeutic option for these cachectic patients. A scoping review was
       conducted to: 1) explore the therapeutic use of cannabis to improve appetite in cancer
       patients, 2) investigate potential reasons for inconsistency amongst available studies, and
       3) identify implications on current practice. Methods: A comprehensive search strategy of
       four electronic databases (MEDLINE, EMBASE, PsycINFO, and Cochrane Central) was
       completed and included the literature published between May 1990 to July 2016. Key
       articles were also hand-searched for further citations, and references from reviews were
       checked to identify additional studies. Results: A total of 315 articles were identified for
       screening, and application of inclusion/exclusion criteria resulted in eight studies being
       included in qualitative synthesis. Small studies (n=6) suggest positive effect between
       tetrohydrocannabinol (THC) use and increased appetite whereas large clinical trials (n=2)
       suggest otherwise. Conclusions: Despite anecdotal observations suggesting the potential
       for cannabis to stimulate appetite, existing studies use various methods of administration
       and dosing, making it difficult to draw meaningful conclusions. Weak methodological
       choices in smaller studies have resulted in a high degree of variability in results. Further
       clinical trials that are well designed and carefully executed are essential to clearly define
       the role of these agents as appetite stimulants.
*
    Correspondence: Carlo DeAngelis, PharmD, RPh, Department of Pharmacy, Sunnybrook Health Sciences Centre,
       2075 Bayview Avenue, Toronto, ON, Canada. E-mail: carlo.deangelis@sunnybrook.ca.
114                      Meiko Peng, Minhaz Khaiser, Michael Lam et al.
                                     INTRODUCTION
The usage of marijuana, also known as cannabis, for medical and recreational purposes has
been documented over thousands of years across different cultures. Asian and Indian historical
literature has described the therapeutic potential of cannabis. Medicinal uses of this agent have
been numerous throughout history and include seizure control, analgesia, anxiolysis, cough
suppression, and appetite stimulation (1, 2).
     Cannabis interacts with the endocannabinoid system (ECS), a ubiquitous lipid signaling
organization found in all vertebrates. Of the two cannabinoid receptors, CB1 (widely
distributed in the central nervous system) is thought to be responsible for behavioral effects,
such as reward, pain perception, and euphoria, while CB2 (found predominantly on B
lymphocytes and natural killer cells) plays a role in immune suppressive actions (3–5). The
purported therapeutic effects of cannabis are due to the activation of CB1 by the main
psychoactive cannabinoid, tetrahydrocannabinol (THC). The isolation of THC from cannabis,
in conjunction with a greater understanding of cannabinoid pharmacology through the
discovery of the ECS, has opened doors to new areas of research, including plausible drug
targets and therapeutic potential in the management of various medical entities such as
chemotherapy-induced nausea and vomiting (6), refractory spasticity in multiple sclerosis (7),
and AIDS-related anorexia (8).
     Many patients with advanced cancer experience decreased appetite and involuntary weight
loss characterized by ongoing loss of skeletal mass that cannot be reversed using conventional
nutritional support, also known as the "cancer anorexia cachexia syndrome" (CACS) (9, 10).
Anorexia (loss of desire to eat) differs from cachexia (progressive wasting of skeletal muscle
mass), but synergistically works with cachexia to reduce food intake and promote weight loss,
thus significantly increasing patient mortality and accounting for up to 20% of cancer deaths
(11). Although the pathophysiology of this syndrome is not yet well defined, the underlying
mechanism is postulated to be secondary to the metabolic changes induced by tumor presence
and growth. These changes are triggered by alterations in the hormonal environment, release
of different tumor factors, and a systemic inflammatory reaction characterized by the
production of pro-inflammatory cytokines, such as interleukins and tumor necrosis factor (12).
CACS continues to be a challenging syndrome to manage, and while optimal therapy involves
treating the underlying cancer, this is not always possible. CACS management requires
integration of both nutritional counseling and pharmacologic agents; however, there are no
established treatment regimens for CACS and options are often limited (13). Megestrol acetate
is currently used as first-line treatment for CACS, and while it can improve the patient’s
appetite and overall weight, it works by increasing fat accumulation with no effect on lean body
mass and quality of life (11, 14). Progestogens such as megestrol are also associated with
significant side effects such as adrenal insufficiency and thrombosis; therefore, given the
possible side effect profile, it is important to explore other possible agents that can optimize
nutritional state and increase quality of life (15, 16).
     Historically, cannabis has been noted to have a significant effect on appetite, which has led
to further exploration into use of the ECS to provide supportive care in patients with CACS
(17–19). It is hypothesized that activation of CB1 receptors regulate appetite at the level of the
hypothalamus, where food intake is controlled, and in the mesolimbic reward system for the
motivational or reward aspects of eating (20, 21). Additionally, stimulation of these receptors
                    Medical marijuana, cancer anorexia and cachexia                            115
on adipocytes and in the liver increases lipogenesis, thus promoting anabolic processes (22).
Nonetheless, the exact mechanism by which cannabinoids exert their effects has yet to be fully
clarified.
     Dronabinol, a synthetic THC analogue, is one of the most commonly studied CB1 agonists.
It was developed initially for the treatment of nausea and vomiting in cancer patients in 1986;
its indication then expanded to also cover treatment of weight loss in HIV-wasting syndrome
(23). At present, there is mixed evidence regarding the use of cannabis as an appetite stimulant
in palliating cancer-associated anorexia. Cannabis is the only antiemetic that may also have
orexigenic properties; however, no clinical trials have been conducted to date that evaluate the
effects of the whole plant on CACS. Furthermore, hindered by political and societal barriers
regarding its legalization status and reputation as an illicit drug, research has been inconsistent
and limited. However, as there are now significant changes in legalization of cannabis, both in
the USA and Canada, this is an opportune time to better understand the effect of this substance
on CACS.
     This scoping review will aim to (1) explore the therapeutic use of cannabis in improving
appetite and related metabolic processes in cancer patients, (2) investigate potential reasons for
inconsistency amongst available studies, and (3) examine implications of available evidence on
current practice.
                                        OUR REVIEW
The current study used a scoping review methodology (24). Key search terms included
neoplasms, cannabis, anorexia and cachexia. Four non-overlapping databases were searched
from May 1990 up to July 2016: Ovid MEDLINE, Ovid Embase Classic, Cochrane Central
Register of Controlled Trials, and PsycINFO. All searches were limited to humans and the
English language (see Table 1). Abstracts that only focused on cachexia or cannabis alone were
excluded, as the purpose of this scoping review is to better understand the effect of cannabis in
CACS. Full text articles were excluded if they were review articles and only focused on
pharmacology to the exclusion of human trials. Key articles were also hand-searched for further
citations for additional relevant studies. Authors then conducted data extraction and appraised
the quality of the included studies.
Table 1. (Continued)
                                           FINDINGS
The search strategy resulted in 315 unique articles (see Figure 1), with 72 full text articles
examined for inclusion/exclusion. After assessing the papers based on the screening criteria,
eight studies that investigated the use of cannabis for appetite stimulation in anorexic/cachectic
cancer patients were identified and all eight were included in the final analysis (see Table 2).
These studies were conducted in the USA (n=4) (21, 25–27), Canada (n=1) (28), Switzerland
(n=1) (29), and Israel (n=1) (30) and published between 1990 and 2015.
                      Medical marijuana, cancer anorexia and cachexia                                117
*focused on cannabis- or cachexia-only, indications other than cachexia, legal processes †review articles,
    pharmacology focused.
Study findings
Author, Year        Study Design     Study        Population                        Intervention                      Appetite              Body Weight
                                     Duration
Sacks et al.        Case report      2 weeks      Patient (n=1) with Hodgkin’s      Intervention: Dronabinol 5 mg     Maintained            Maintained
1990 (31)                                         disease                           three times daily
                                                  Age: 60                           Control: N/A
                                                  Active treatment
Plasse et al.       Dose-ranging     3-6 weeks    Adult cancer patients (n=42);     Intervention arm 1: Dronabinol     2.5 mg twice daily   Maintained at all
1991 (25)           pilot study                   most common tumor types were      2.5 mg daily                      group (Increased      dose levels
                                                  lung, prostate, colon             Intervention arm 2: 2.5 mg        47%)
                                                  Mean age: not reported            twice daily
                                                  Active treatment: one course of   Intervention arm 3: 5 mg daily
                                                  chemo permitted during study      Intervention arm 4: 5 mg twice
                                                  period                            daily
                                                                                    Control: N/A
Nelson et al.       Phase II,        4 weeks      Adult patients (n=19) with        Intervention: Dronabinol 2.5      Increased             Not measured
1994 (26)           prospective,                  cancer-associated anorexia Life   mg three times daily one hour
                    open label                    expectancy >4 weeks               after meals
                    study                         Mean age: not reported            Control: N/A
                                                  Active treatment: N/A
Jatoi et al. 2002   Phase III        Megestrol    Adult advanced cancer patients    Intervention arm 1: Megestrol     Megesterol            Megestrol
(27)                clinical trial   (80 days),   (n=469) with CACS (≥5 pounds      acetate 800 mg/d liquid           (Increased 75%)       (Increased 11%)
                                     dronabinol   weight loss during preceding 2    suspension plus placebo           Dronabinol            Dronabinol
                                     (57 days),   months and/or a daily intake of   Intervention arm 2: Dronabinol    (Increased 49%)       (Increased 3%)
                                     combo (74    ≤20 calories/kg of body weight)   2.5 mg twice daily plus           Combo                 Combo
                                     days)        Life expectancy ≥ 3 months        placebo                           (Increased 66%)       (8%)
                                                  Mean age: 66                      Intervention arm 3: both
                                                  Active treatment: chemo or        agents above
                                                  radiation permitted
Walsh et al.        Case series      2-30         Adult cancer patients (n=6)       Intervention: Dronabinol 2.5      Increased (50%)       Increased (67%)
2005 (21)                            weeks        Mean age: N/A                     mg three times daily for 4
                                                  Active treatment: N/A             weeks (26). Patients then given
                                                                                    opportunity to escalate dose if
                                                                                    tolerated without toxicity. Max
                                                                                    15 mg daily.
                                                                                    Control: N/A
                                                                                                                                                     119
Author, Year      Study Design     Study      Population                        Intervention                     Appetite             Body Weight
                                   Duration
Strasser et al.   Phase III        6 weeks    Adult advanced cancer patients    Intervention arm 1: Cannabis     Cannabis extract     Maintained
2006 (29)         clinical trial              (n=243) with CACS (weight         extract capsule (2.5 mg THC      (Increased 73%)
                                              loss ≥5% over 6 months)           and 1 mg cannabidiol) twice      Dronabinol
                                              Life expectancy of 3 months       daily                            (Increased 58%)
                                               and ECOG ≤2                      Intervention arm 2: Dronabinol   Placebo (Increased
                                              Mean age: 61                      2.5 mg twice daily               69%)
                                              Active treatment                  Control: placebo orally twice
                                                                                daily
Brisbois et al.   Phase II         22 days    Adult advanced cancer patients    Intervention: Dronabinol 2.5     Increased (64%)      Not measured
2011 (28)         pilot trial                 (n=21) with poor appetite and     mg twice daily
                                              chemosensory alterations          Control: Placebo oral capsules
                                              (decreased food intake ≥2         twice daily
                                              weeks)
                                              Life expectancy >2 months
                                              Mean age: 67
                                              Active treatment: chemo
                                              permitted
Waissengrin et    Observational    1 year     Adult patients (n=279), treated   None; measured perceptions of    Increased (60%)      Not measured
al. 2015 (30)     study                       and received permit for medical   patients regarding efficacy of
                                              cannabis                          cannabis
                                              Mean age: 60
                                              Active treatment
120                      Meiko Peng, Minhaz Khaiser, Michael Lam et al.
     Anorexia and cachexia are common in palliative care populations and are independent risk
factors for morbidity and mortality (36). The first investigation of using cannabis as an appetite
stimulant in cancer patients was conducted by Regelson et al. in 1975 (37). In this Phase II,
placebo-controlled, cross-over study of 54 patients with advanced cancer, significant weight
gain was observed with three oral doses of 0.1 mg/kg/day of dronabinol. However, almost 40%
of patients discontinued treatment due to side effects (drowsiness, mood changes, and memory
problems), perhaps indicating that the majority of patients would not be able to tolerate higher
doses in order to realize the beneficial effects on appetite (37). A case report suggested benefit
of 5 mg dronabinol taken orally three times daily in a patient with Hodgkin’s lymphoma by
maintaining adequate food intake during chemotherapy (31). Other Phase II studies in the 1990s
also suggest cannabinoids’ potential at fixed doses of 2.5 mg THC two to three times daily in
advanced cancer patients (25, 26). Both studies noted that THC was effective and well-tolerated
at low doses. Despite these positive results, the studies are limited by high dropout rates,
variable patient inclusion criteria, placebo effect, and small sample sizes. For example, in the
case report (31), the use of a placebo was not possible and the results were obtained via a self-
recorded food diary. Moreover, Nelson et al.’s Phase II clinical trial was also not blinded or
controlled, and enrolled only 19 patients over 4 weeks (26). Although they found a reduction
in anorexia in 68% of patients, only 53% were able to complete the 28-day trial, and 16% had
to suspend the treatment due to intervention-associated toxicity. Taken together, these trials
suggest a modest benefit, associated with significant toxicity, from dronabinol. It is important
to note that most orally-dosed studies use dronabinol, THC formulated in sesame oil and
supplied as soft gelatin capsules. In comparison, inhalation studies use the dried plant that
contains other potentially active cannabinoids other than THC alone.
     Fortunately, trial robustness and scientific rigor improved moving into the 21st century.
Prompted by the positive findings of previous Phase II trials, further investigations were made
into the cannabinoids. A Phase III trial of 469 advanced cancer patients assessed the efficacy
of low-dose dronabinol (2.5 mg twice daily) against high-dose megestrol (800 mg daily) in
palliating cancer-associated anorexia (27). Despite the previously positive results, the study
showed that dronabinol was inferior to megestrol, with only 49% versus 75% of patients noting
an improved appetite. A combination of both treatments did not appear to confer additional
benefit, suggesting that the capacity for appetite stimulation is easily saturated. Further
investigation was undertaken by a Swiss group who performed a Phase III trial that compared
the weight-inducing effects of cannabis extract (1:1 ratio of THC:CBD) or dronabinol to
placebo in 243 patients with CACS. Results showed no improvement in appetite or quality of
life in the treatment group (29). These two large multicenter clinical trials presented very
different results as what one would expect based on the smaller studies previously conducted.
     Recently, in a twelve-month study period of non-cancer patients who failed to respond to
treatment for anorexia and weight loss, dronabinol use was associated with a trend toward
weight gain (38). Minor weight gain was also observed in an older population with Alzheimer’s
disease (39). Furthermore, a randomized, double-blinded, placebo-controlled pilot study
suggested that cancer patients with altered chemosensory had increased appetite and improved
taste with dronabinol (2.5 mg twice daily) compared to placebo (40). Though this was a small
and short trial, with only 21 patients completing the study over 18 days, the goal of the study
was for proof-of-concept and to provide a starting point for future trials to build upon. There is
increasing interest in other forms of medical marijuana, such as by inhalation or ingestion of
oils from natural legal sources; however, no clinical trials have tested the efficacy of these
                    Medical marijuana, cancer anorexia and cachexia                          121
formulations in cancer patients to date. An Israeli study analyzed the indication of cannabis in
cancer patients who are actively being treated with the agent (>90% via smoking) (30). Of the
multiple indications for which cannabis was prescribed, anorexia followed pain as the second
most cited (56%), and was reported to improve appetite in 60% of patients (30).
                                       DISCUSSION
With cannabis quickly gaining prominence in the medical world for its therapeutic potential,
several trials have focused on supportive care issues in cancer; however, clinical trials for
CACS are difficult to conduct and become even more challenging to interpret when trying to
establish clinical practice guidelines.
Route of administration
The two main routes of administration studied are oral extracts of THC analogues (tablets)
and inhalation of marijuana (cigarettes). It is interesting to note that while all smoking
studies, which were conducted in non-cancer populations, reported a positive effect of cannabis
on appetite (33–35), oral studies seem to be inconsistent, with the two largest clinical trials
demonstrating negative findings (27, 29). Despite the multiple smoking studies that
demonstrated positive correlations between cannabis and appetite before the 21st century (33,
34), adverse effects of smoking have since then gained prominence in the medical community.
Consequently, modern clinical trials are wary of utilizing this method due to the many
documented toxic effects associated with smoking. This presents a dilemma; while smoking is
the predominate method used in anecdotal reports of cannabis-induced appetite, use of smoking
is associated with clear harm and as such has not been used in CACS studies. However, recent
studies show promise for vaporization (heating to a point where cannabinoids are emitted
without combustion) due to not only faster onset without negative effects of smoking but also
ability for self-regulation of dosage (41). Smoking is the most common method of cannabis
ingestion, as the dried plant is often conveniently rolled into a cigarette-type vehicle. Though
the pharmacologic and psychoactive cannabinoids are found in all parts of the plant, they are
traditionally accessed from the dried leaves and flowers of the hemp plant (42). The THC
content varies considerably depending on the strain of cannabis, but typically ranges from 5-
25% of the dry plant weight (41). Since the dried plant contains many other potentially active
cannabinoids that can account for activities not observed with THC alone, these constituents
may also interact with specific cannabinoid receptors to modulate the hyperphagic system. The
intervention used in oral studies contained only THC, thus it is inappropriate to extrapolate and
compare the results of THC-only versus dried plant studies.
     Inhalation achieves rapid drug delivery from the lungs to the brain and studies have shown
that more than 2000 diverse compounds are produced by pyrolysis during smoking of cannabis
(43). Multiple animal studies suggest that exposure to cannabis smoke alone has
pharmacological consequences (44). In comparison, the bioavailability of oral intake is low and
variable primarily due to the extensive first pass metabolism. Furthermore, oral intake can be
inconsistent for different people due to inter-dependent patient factors. When THC passes
122                      Meiko Peng, Minhaz Khaiser, Michael Lam et al.
through the liver, it is metabolized into an equipotent hydroxylated product, which may be more
psychoactive for some people depending on individual cytochrome P450 enzyme activity. Thus
while most patients have a slower effect from oral intake, some patients report a more
significant psychoactive effect compared with those who inhale it. There are many options for
oral intake aside from the capsule form, such as edibles (in form of baked goods) and THC-
infused oils. Studies have shown that baked products yield low and irregular plasma
concentrations and thus are not practical to use in research studies (45). On the other hand, the
oil formulation presents as a viable avenue for research because administration of the oil
without the gelatin capsule may increase bioavailability and provide patients with similar
benefits as smoking without the pulmonary risks. Regardless, the myriad of discrepancies
between routes of administration, such as patient dependent factors and pyrolysis byproducts,
may help explain the inconsistencies observed in inhalation versus oral studies.
Although the inclusion criteria for patients in recent studies are more explicitly defined, they
remain inconsistent between studies, making them difficult to compare. Inclusion of patients
with metastatic and incurable cancer versus those of potentially curable malignancies may
yield significantly different results. Studies have shown that nutritional counselling leads to
improvement in cancer symptoms and quality of life, but not in chemotherapy-treated patients
with advanced disease (46). Thus since nutritional counselling is often standard of care when
treating CACS, depending on the disease progression, the patient may respond differently.
Most studies also did not specify the degree and intensity of nutritional counseling, potentially
having a significant impact on results. It is unknown whether cannabis’ effectiveness as an
appetite stimulant is dependent on the severity of the disease, and a greater focus on better
defined patient populations is required to tease out these discrepancies.
     Concurrent chemotherapy or radiation therapy are also important factors for consideration.
Some patients may do better on concurrent treatment, and derive these benefits in the form of
increased appetite and weight gain (47). Therefore, studies should balance treatment groups on
the basis of whether the patients were receiving concomitant cancer therapy. For example, in
the phase III trial that compared dronabinol against megestrol in patients with advanced cancer,
chemotherapy or radiation was permitted throughout the study period (27). Thus it is unclear
whether any observed weight gain was due to the intervention or as a result of concurrent anti-
cancer therapy that reduced tumor burden and thus CACS.
     Finally, the criteria for cancer-associated weight loss remain questionable as each study set
their own defined measures, whether noting a numerical target of weight loss in pounds, or as
a percentage weight loss. To date, it remains unclear as to the optimal point at which anti-
cachectic treatment should be introduced. As CACS is a continuum, it is difficult to both assess
the stage of CACS the patient falls into, and decide when treatment should start. Consequently,
efforts have been made to develop a framework for the definition and classification of cancer
cachexia. An international consensus publication was published in 2011 that defines the
diagnostic criteria for various stages of cancer cachexia. Cachexia is predominantly defined as
weight loss >5% over the past 6 months (in absence of simple starvation); or BMI <20 and any
degree of weight loss >2% (9). Only one study recruited patients based on these consensus
criteria (29). At present, further studies are required to standardize the patient inclusion process
                    Medical marijuana, cancer anorexia and cachexia                            123
and assess patients at the same point of the CACS spectrum in order to draw meaningful
conclusions on the therapeutic potential of cannabis.
Dosing inconsistencies
The effects of dronabinol are dose-dependent as studies have shown only specific doses are
associated with appetite stimulation (25). Jatoi and colleagues’ study presented a negative
correlation between dronabinol (2.5 mg twice daily) and appetite, but this result was criticized
for its use of low dose THC, potentially impacting its ability to demonstrate superiority over
megestrol (48). However, the authors justified their dosing choice based on results from a
previous study which showed higher toxicity rate with higher doses of dronabinol (26).
Nevertheless, it is difficult to interpret Jatoi et al.’s phase III trial findings because they
evaluated dronabinol at a dose of 2.5 mg twice daily, while Nelson et al.’s phase II trial assessed
a dose of 2.5 mg three times daily. Similarly, Strasser et al.’s negative findings in the large
Phase III trial were also criticized on their low dosing selections. The incidence of adverse
events was similar for the treatment group versus placebo; this either confirms dronabinol’s
safety profile, or suggests that the chosen dose was too low to demonstrate efficacy.
Unfortunately, there are few dose titration trials of dronabinol in cancer patients that can
establish an optimal dose; further research is required in this area (37, 49).
Anorexia and cachexia are common symptoms in palliative care populations, and the use of
cannabis as an appetite stimulant in other chronic disease have also been explored. Cannabis
has been advocated in patients with HIV/AIDS in order to increase appetite, promote weight
gain, and improve mood (50). In placebo-controlled studies of HIV-positive chronic cannabis
smokers, smoking cannabis led to an increase in food intake (51–53). Dronabinol has been
studied in treatment of HIV wasting, and has been approved for the treatment of this syndrome
(41, 49). Akin to CACS, proinflammatory cytokines are likely the major factor responsible for
HIV wasting (54). Although dronabinol has had positive effects in studies for HIV cachexia,
these effects have not been replicated in the cancer population.
    The evidence for cannabis’ positive effects for HIV-wasting syndrome are limited, and was
largely based upon a single placebo-controlled trial that demonstrated dronabinol significantly
increased appetite in 139 AIDS patients, even with long-term use (55, 56). Even though
monotherapy with dronabinol was associated with appetite stimulation, when compared head-
to-head with megestrol in a small randomized trial of 39 patients with HIV-wasting syndrome,
dronabinol was again found to be inferior and combination therapy did not confer additional
benefits (57). These results mirror those found in the dronabinol versus megestrol study
amongst cancer patients (27). Criticisms of early positive observations in HIV patients include
small number of participants and inadequate length of study that focused only on short-term
effects. After expanding its indication to include treatment of AIDS-related cachexia in 1992,
dronabinol was discontinued in 2012 in Canada by the manufacturer (41). Overall, similar to
the cancer literature, there is currently insufficient research into the use of dronabinol for
124                     Meiko Peng, Minhaz Khaiser, Michael Lam et al.
appetite stimulation in HIV wasting, and long-term data showing a sustained effect in this
population has yet to be presented (58).
Observations of increased appetite from acute cannabis use has set the stage for studies in
patients with HIV and cancer, and in both clinical venues, marijuana has been purported to
stimulate appetite with varying strengths of evidence. However, recent studies indicate that
while appetite may be stimulated, the weight gain may not be clinically meaningful. This was
first alluded to when Strasser et al. failed to find a difference in weight gain in the treatment
group (29). According to several large epidemiological studies, they found that cannabis use in
healthy populations is associated with a lower rather than a higher body mass index, despite
consuming increased calories (59–61). For example, in a 15-year longitudinal study with 3,617
participants, the researchers found that cannabis users had higher daily calorie intake but no
increase in body mass index (59). These puzzling results challenge the traditional notion that
cannabis causes weight gain, and suggest that the effects of cannabis is a function of initial
weight status (62). This may contribute to the inconsistent results noted from both the HIV and
cancer literature, as no standardization was made based on initial patient weight. Furthermore,
there could be differences between short-term and chronic cannabis use as the longest study to
date only looked at 7 months of treatment. The type of weight gain is also an important
assessment because to date, no treatment options have assessed and distinguished weight gain
in terms of fat accumulation or increases in lean body mass. Future studies should consider
incorporating bioelectrical impedance analysis that can measure body composition to assess
whether weight gain with dronabinol is caused by fat, lean body mass, or water (63).
Cancer anorexia and cachexia continues to be responsible for up to one out of five deaths in
cancer patients, and is considered an important comorbidity of cancer. Current guidelines
classify the syndrome into three stages of clinical relevance: pre-cachexia, cachexia, and
refractory cachexia. Considered as a dynamic process, it is likely that the progression of CACS
may be modulated by timely and effective interventions with nutrition and/or pharmacological
therapy. Indeed, studies have shown that in head/neck, lung and colorectal cancer patients, early
intervention may maintain nutritional status and improve clinical outcome (64–66).
     Appetite stimulants such as anti-inflammatory agents and anabolic steroids are some of the
current and emerging treatment options for CACS. However, no single pharmacological agent
has shown clear efficacy in counteracting depletion of muscle mass and in attenuating CACS
symptoms (67). There continues to be a lack of consensus in clinical application of cannabinoid
in cancer anorexia and cachexia. Currently, the only international guideline available has been
published by the French National Federation of Cancer Centers working group on the use of
appetite stimulants in oncology patients (68). The current first and second line treatments,
megestrol and corticosteroids, respectively, are recommended with level B evidence (good
quality evidence from randomized trials and results are consistent when considered together).
In agreement with our findings, dronabinol was found to have level C evidence, indicating that
                       Medical marijuana, cancer anorexia and cachexia                                       125
the methodology of available studies is weak and that the results when taken together are not
consistent. Despite the lack of convincing evidence, the Marijuana for Medical Purposes
Regulation (MMPR) currently permits the use of cannabis for cancer-related cachexia where
other agents have failed, and select patients continue to find benefits for appetite stimulation
with the medication.
Study limitations
Though studies were assessed for quality and the results helped identify parameters and gaps
in the available literature, a detailed data extraction and quantitative synthesis was not
performed. Also, there is no guarantee that all cannabis interventions in CACS were retrieved
as a result of the limitation using MeSH terms. This may have contributed to the low number
of results obtained, and perhaps a more comprehensive search strategy could have generated
further insight. Moreover, this review identified studies that used synthetic THC (dronabinol)
instead of cannabis as the intervention; there may be differences in outcomes between herbal
cannabis and synthetic THC which could not be assessed in this study. Finally, as there is a
dearth of studies in humans, future syntheses may consider including animal studies in order to
increase the scope of the review and to better understand how cannabis could affect CACS.
                                             CONCLUSION
The progress achieved over the past decade in understanding the ECS has revived the
therapeutic interest in cannabis. It is clear that this system plays a role in diverse biological
pathways, including modulation of appetite. Historical and anecdotal observations suggest the
potential of cannabis as an appetite stimulant, but existing studies are littered with
inconsistencies in terms of standardization of patient inclusion criteria, route of administration,
and dosing, making it difficult to draw meaningful conclusions across trials. Specifically, in the
setting of CACS, future well-designed clinical trials that are carefully executed are essential to
clearly define the role of these agents as appetite stimulants. Although cannabis’ anti-emetic
and orexigenic effects suggest potential benefit in the palliative care setting for patients with
cancer, there are currently no data from controlled studies to indicate a significant benefit from
ingested marijuana extract or smoked marijuana for cancer-related anorexia and/or cachexia.
                                             REFERENCES
[1]   Lemberger L. Potential therapeutic usefulness of marihuana. Ann Rev Pharmacol Toxicol 1980;20:151–
      72.
[2]   O’Shaughnessy WB. On the preparations of the Indian hemp, or Gunjah: Cannabis indica their effects
      on the animal system in health, and their utility in the treatment of tetanus and other convulsive diseases.
      Prov Med J Retrosp Med Sci 1843;5(123):363.
126                         Meiko Peng, Minhaz Khaiser, Michael Lam et al.
[28]   Brisbois TD, de Kock IH, Watanabe SM, Mirhosseini M, Lamoureux DC, Chasen M, et al. Delta-9-
       tetrahydrocannabinol may palliate altered chemosensory perception in cancer patients: results of a
       randomized, double-blind, placebo-controlled pilot trial. Ann Oncol 2011;22(9):2086–93.
[29]   Strasser F, Luftner D, Possinger K, Ernst G, Ruhstaller T, Meissner W, et al. Comparison of orally
       administered cannabis extract and delta-9- tetrahydrocannabinol in treating patients with cancer-related
       anorexia-cachexia syndrome: a multicenter, phase III, randomized, double-blind, placebo-controlled
       clinical trial from the Cannab. J Clin Oncol 2006;24(21):3394–400.
[30]   Waissengrin B, Urban D, Leshem Y, Garty M, Wolf I. Patterns of use of medical cannabis among Israeli
       cancer patients: a single institution experience. J Pain Symptom Manage 2015;49(2):223–30.
[31]   Sacks N, Hutcheson JR, Watts JM, Webb RE. Case report: the effect of tetrahydrocannabinol on food
       intake during chemotherapy. J Am Coll Nutr 1990;9(6):630–2.
[32]   Hollister LE. Hunger and appetite after single doses of marihuana, alcohol, and dextroamphetamine.
       Clin Pharmacol Ther 1971;12(1):44–9.
[33]   Foltin RW, Fischman MW, Byrne MF. Effect of smoked marijuana on food intake of humans living in
       a residential laboratory. Appetite 1988;11:1–14.
[34]   Greenberg I, Kuehnle J, Mendelson JH, Bernstein JG. Effects of marihuana use on body weight and
       caloric intake in humans. Psychopharmacology (Berl) 1976;49(1):79–84.
[35]   Mattes RD, Engelman K, Shaw LM, Elsohly MA. Cannabinoids and appetite stimulation. Pharmacol
       Biochem Behav 1994;49(1):187–95.
[36]   Davis MP, Dreicer R, Walsh D, Lagman R, LeGrand SB. Appetite and cancer-associated anorexia: a
       review. J Clin Oncol 2004;22(8):1510–7.
[37]   Regelson W, Butler J, Schultz J. Tetra-hydrocannbinol as an effective antidepressant and appetite-
       stimulating agent in advanced cancer patients. Int Conf Pharmacol Cannabis. 1975;763–76.
[38]   Wilson M, Philpot C, Morley J. Anorexia of aging in long term care: is Dronabinol an effective appetite
       stimulant? A pilot study. J Nutr Health Aging 2007;11(2):195–8.
[39]   Volicer L, Stelly M, Morris J, McLaughlin J, Volicer BJ, Rogers EN. Effects of dronabinol on anorexia
       and disturbed behavior in patients with Alzheimer's disease. Int J Geriat Psychiatry 1997;12(9):913–9.
[40]   Brisbois TD, de Kock IH, Watanabe SM, Mirhosseini M, Lamoureux DC, Chasen M, et al. Delta-9-
       tetrahydrocannabinol may palliate altered chemosensory perception in cancer patients: results of a
       randomized, double-blind, placebo-controlled pilot trial. Ann Oncol 2011;22(9):2086–93.
[41]   Abramovici H. Information for Health Care Professionals: Cannabis (marijuana) and the cannabinoids.
       Health Canada 2013. URL: http://www.hc-sc.gc.ca/dhp-mps/marihuana/med/infoprof-eng.php.
[42]   Ben Amar M. Cannabinoids in medicine: a review of their therapeutic potential. J Ethnopharmacol
       2006;105(1-2):1–25.
[43]   Sharma P, Murthy P, Bharath MMS. Chemistry, metabolism, and toxicology of cannabis: clinical
       implications. Iran J Psychiatry 2012;7(4):149–56.
[44]   Lichtman AH, Poklis JL, Poklis A, Wilson DM, Martin BR. The pharmacological activity of inhalation
       exposure to marijuana smoke in mice. Drug Alcohol Depend 2001;63:107–16.
[45]   Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers 2007;4(8):1770–804.
[46]   Ovesen L, Allingstrup L, Hannibal J, Mortensen EL, Hansen OP. Effect of dietary counseling on
       food intake, body weight, response rate, survival, and quality of life in cancer patients undergoing
       chemotherapy: a prospective, randomized study. J Clin Oncol 1993;11(10):2043–9.
[47]   Giordano KF, Jatoi A. The cancer anorexia/weight loss syndrome: therapeutic challenges. Curr Oncol
       Rep 2005;7:271–6.
[48]   Reynolds R. Comparative efficacy of dronabinol and megestrol acetate. Am Soc Clin Oncol
       2002;20(12):2912–3.
[49]   Struwe M, Kaempfer SH, Geiger CJ, Pavia AT, Plasse TF, Shepard K V, et al. Effect of dronabinol on
       nutritional status in HIV infection. Ann Pharmacol 1993;27:827–31.
[50]   Woolridge E, Barton S, Samuel J, Osorio J, Dougherty A, Holdcroft A. Cannabis use in HIV for pain
       and other medical symptoms. J Pain Symptom Manage 2005;29(4):358–67.
[51]   Haney M, Rabkin J, Gunderson E, Foltin RW. Dronabinol and marijuana in HIV+ marijuana smokers:
       Acute effects on caloric intake and mood. Psychopharmacology (Berl) 2005;181:170–8.
128                         Meiko Peng, Minhaz Khaiser, Michael Lam et al.
[52]   Haney M, Gunderson EW, Rabkin J, Hart CL, Vosburg SK, Comer SD, et al. Dronabinol and marijuana
       in HIV-positive marijuana smokers: caloric intake, mood, and sleep. J Acquir Immune Defic Syndr
       2007;45(5):545–54.
[53]   Abrams DI, Hilton JF, Leiser RJ, Shade SB, Elbeik TA, Aweeka FT, et al. Short-term effects of
       cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial. Ann
       Intern Med 2003;139(4):258-66.
[54]   Morley JE, Thomas D, Wilson M. Cachexia: pathophysiology and clinical relevance. Am Soc Nutr
       2006;83:735–43.
[55]   Beal JE, Olson R, Laubenstein L, Morales JO, Bellman P, Yangco B, et al. Dronabinol as a treatment
       for anorexia associated with weight loss in patients with AIDS. J Pain Symptom Manage 1995;10(2):89–
       97.
[56]   Beal JE, Olson R, Lefkowitz L, Laubenstein L, Bellman P, Yangco B, et al. Long-term efficacy and
       safety of dronabinol for acquired immunodeficiency syndrome-associated anorexia. J Pain Symptom
       Manage 1997;14(1):7–14.
[57]   Timpone J, Wright D, Li N, Egorin M, Enama M, Mayers J, et al. The safety and pharmacokinetics of
       single-agent and combination therapy with megestrol acetate and dronabinol for the treatment of HIV
       wasting syndrome. The DATRI 004 Study Group. Division of AIDS Treatment Research Initiative.
       AIDS Res Hum Retroviruses 1997;13(4):305-15.
[58]   Lutge EE, Gray A, Siegfried N. The medical use of cannabis for reducing morbidity and mortality in
       patients with HIV/AIDS. Cochrane Database Syst Rev 2013;(4):CD005175.
[59]   Rodondi N, Pletcher MJ, Liu K, Hulley SB, Sidney S; Coronary Artery Risk Development in Young
       Adults (CARDIA) Study. Marijuana use, diet, body mass index, and cardiovascular risk factors (from
       the CARDIA study). Am J Cardiol 2006;98(4):478–84.
[60]   Le Strat Y, Le Foll B. Obesity and cannabis use: results from 2 representative national surveys. Am J
       Epidemiol 2011;174(8):929–33.
[61]   Hayatbakhsh MR, O’Callaghan MJ, Mamun AA, Williams GM, Clavarino A, Najman JM. Cannabis
       use and obesity and young adults. Am J Drug Alcohol Abuse 2010;36(6):350-6.
[62]   Sansone RA, Sansone LA. Marijuana and body weight. Innov Clin Neurosci 2014;11(7-8):50–4.
[63]   Toso S, Piccoli A, Gusella M, Menon D, Crepaldi G, Bononi A, et al. Bioimpedance vector pattern in
       cancer patients without disease versus locally advanced or disseminated disease. Nutrition 2003;19
       (6):510–4.
[64]   Paccagnella A, Morello M, Da Mosto MC, Baruffi C, Marcon ML, Gava A, et al. Early nutritional
       intervention improves treatment tolerance and outcomes in head and neck cancer patients undergoing
       concurrent chemoradiotherapy. Support Care Cancer 2010;18(7):837–45.
[65]   van der Meij BS, Langius JAE, Spreeuwenberg MD, Slootmaker SM, Paul MA, Smit EF, et al. Oral
       nutritional supplements containing n-3 polyunsaturated fatty acids affect quality of life and functional
       status in lung cancer patients during multimodality treatment: an RCT. Eur J Clin Nutr 2012;66(3):399–
       404.
[66]   Chen Y, Liu BL, Shang B, Chen AS, Liu SQ, Sun W, et al. Nutrition support in surgical patients with
       colorectal cancer. World J Gastroenterol 2011;17(13):1779–86.
[67]   Mantovani G, Maccio A, Madeddu C, Serpe R, Massa E, Dessi M, et al. Randomized phase III clinical
       trial of five different arms of treatment in 332 patients with cancer cachexia. Oncologist 2010;15
       (2):200–11.
[68]   Desport JC, Blanc-Vincent MP, Gory-Delabaere G, Bachmann P, Béal J, Benamouzig R, et al.
       Standards, Options and Recommendations (SOR) for the use of appetite stimulants in oncology. Work
       group. Federation of the French Cancer Centres (FNCLCC). Bull Cancer 2000;87(4):315–28.
In: Cannabis: Medical Aspects                                             ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                    © 2017 Nova Science Publishers, Inc.
Chapter 11
       In Canada, patients may access dried/fresh cannabis or cannabis oil from licensed
       producers when prescribed for medical use. Pain management is the most common reason
       for medical cannabis use and strategies for dosing, potency, and selection of modes of
       delivery need to be further explored. This scoping review aimed to determine therapeutic
       dosing strategies for different forms of cannabis in acute or chronic pain conditions. A
       literature search was performed in four databases – MEDLINE, Embase, Cochrane Central
       and PsycInfo. Additional publications were identified by hand searching key articles,
       reviews and Health Canada documents. In total, 11 articles were selected for inclusion.
       Seven randomized controlled trials assessed smoked cannabis use in different pain
       conditions: three studies used vaporized cannabis; and one retrospective, observational
       study reviewed different cannabis formulations. Current literature supporting
       smoked/vaporized cannabis is limited by small sample sizes, short intervention periods,
       and large variability in dosing. Evidence for oral forms of cannabis (oils, brownies) is even
       more limited. Vaporizing should be recommended over smoking cannabis and the best
       dosing strategy is to start at a low delta-9-tetrahydrocannabinol (THC) potency (e.g., 1%
       THC) and slowly titrate to tolerance at 3-4 inhalations daily with ~45 seconds between
       inhalations. The same strategy can be used with oils while maintaining at least two hours
       between individual doses to assess pain relief versus side effect profile.
*
    Correspondence: Carlo DeAngelis, PharmD, RPh, Department of Pharmacy, Sunnybrook Health Sciences Centre,
       2075 Bayview Avenue, Toronto, ON, Canada. E-mail: carlo.deangelis@sunnybrook.ca.
130                      Minhaz Khaiser, Meiko Peng, Michael Lam et al.
                                     INTRODUCTION
Cannabis, also called marijuana (derived from the plant Cannabis sativa or indica), is one of
the most popular recreational drugs used worldwide and has a long history of medicinal use
dating as far back as 2737 B.C. (1, 2). The plant material contains many different classes of
compounds and more than 70 phytocannabinoids that interact with receptors of the
endocannabinoid system (ECS). The ECS is a ubiquitous lipid signaling system containing the
cannabinoid 1 and 2 receptors (CB1 and CB2) that regulate various immune and nervous
system processes in the body. These include inflammation, neural development and pain (1, 3,
4). The most well studied cannabinoid, THC, is associated most with the physical and
psychotropic effects of cannabis (5). The second most abundant cannabinoid, cannabidiol
(CBD), is minimally psychoactive and present in lesser amounts (4). Thus, medical cannabis is
available as different strains primarily classified by potency or percentage of THC available.
     Medical use of “cannabis” in its dried plant or oil form will be the focus of this review.
This term should not be confused with “cannabinoids”, which refers to the synthetic forms of
cannabis where one or two active molecules have been extracted from the plant material to
create medications commercially available for oral use: nabilone (synthetic analogue of THC,
Cesamet®), dronabinol (synthetic THC, Marinol®), or nabiximols (oromucosal spray,
Sativex®; containing THC:CBD in approximately equal amounts) (4, 6).
     Cannabis is commonly available as dried herb, oil, or butter (4, 6). There are also synthetic
formulations available in the form of capsules (containing sesame oil) and plant extracts as
oromucosal sprays (nabiximols/Sativex®) (4). It can be administered orally by mixing with
foods, infused in tea, inhaled by smoking or vaporizing, or rarely administered topically or
rectally (4, 6). Of the available modes of delivery, smoking is the most common and preferred
form as it results in a more rapid onset of action and delivery of THC to the brain compared to
oral consumption (7, 8). Smoking cannabis is the act of inhaling smoke produced by burning
the plant material in hand-rolled cigarettes or pipes with temperatures for combustion reaching
200° Celsius (C) or higher (7, 9). Vaporizing is an increasingly popular alternative to smoking
where the herb is heated below the combustion point so that the vapors produced lack hazardous
compounds such as tar, carbon monoxide and carbon dioxide present in cannabis smoke (7).
Vaporizing and smoking have comparable onsets of action (10).
     With growing evidence to prove that targeting the ECS can have therapeutic benefits,
cannabis is being used to treat a wide variety of conditions such as nausea and vomiting,
cachexia and appetite stimulation, and pain. Cannabinoid receptors of the ECS have been found
in many areas of the central and peripheral pain pathways and several mechanisms have been
associated with analgesic effects of cannabis (11, 12). These include antinociceptive effects in
descending pain pathways, inhibition of prostaglandin synthesis reducing inflammation, and
modulation of neural activity centrally (1, 13). Some studies have shown that concomitant use
of THC can enhance analgesic effects of morphine by increasing activation of kappa and delta
opioid receptors (14). Cannabis has been prescribed commonly for pain related conditions such
as neuropathic pain, musculoskeletal pain, chemotherapy-induced pain and Multiple Sclerosis
(MS) related refractory pain (15). Systematic reviews of randomized trials have shown
supporting evidence of cannabis and cannabinoid use in various pain conditions, but have not
provided clear dosing recommendations (15–17). With the increase in chronic pain related
               Medical cannabis dosing strategies in pain related conditions                   131
                                        OUR REVIEW
A scoping review methodology was used to quickly explore existing literature on the topic.(21)
Eligible studies were identified using four non-overlapping databases including MEDLINE,
Embase, Cochrane Central and PsycInfo. The search strategy included the following broad
terms: cannabis, cannabinoid, pain, and dose-response relationship (see Table 1). Additional
papers were identified through hand searching of key articles, reviews and government (Health
Canada) documents.
     Initially, the titles and abstracts were subject to the following inclusion criteria: cannabis
use was evaluated in a pain management setting; formulation of cannabis used included
smoking, vaporizing, topicals, edibles (cookies, brownies, teas, infusions, butters or oils);
studies were conducted in humans; studies were published in English; and the full article was
retrievable. Studies were excluded if they: solely used synthetic cannabinoids (dronabinol,
nabilone and other novel cannabinoids) or oromucosal sprays of cannabis extracts; did not
assess effect of cannabis on pain; used animal models; reviewed the pharmacology of cannabis;
and full text was inaccessible. The first author conducted initial data extraction (see Table 2).
Extracted data included study design, participant characteristics (age, type of pain), sample size,
intervention (type, dose, duration), and clinical outcomes.
                                          FINDINGS
The search strategy resulted in 900 potentially eligible articles and 5 other articles were found
through review of references, including the Information for Health Care Professionals
document published by Health Canada (see Figure 1) (4). After applying eligibility criteria and
excluding duplicates, a full-text review of 44 articles was performed. Eleven publications in
total were selected for inclusion in this scoping review (see Table 2). Studies were conducted
in America (n = 9 studies) (22–30) and Canada (n = 2 studies) (31, 32) and were published
between 2007 and 2016. All studies that solely used smoked or vaporized cannabis had a
randomized, double-blind and placebo controlled design. The intervention potencies used in all
studies were represented as percentage of THC by weight of cannabis material. As smoked and
132                       Minhaz Khaiser, Meiko Peng, Michael Lam et al.
vaporized forms of cannabis were the two most common interventions, the relevant studies
have been discussed in detail below.
In order to standardize dosing of inhaled forms of cannabis, a common, modified smoking and
vaporizing inhalation protocol was implemented called the “Foltin Puff Procedure” (33). The
standardized protocol requires participants to inhale for 5 seconds, hold inhalation for 10
seconds, and then exhale fully. Participants were also required to have 40-45 seconds resting
periods between inhalations. Subjects were mostly given verbal cues by investigators or clinical
trial nurses (23, 24, 29), or were taught to self-administer at home using a titanium pipe in one
case (32).
                              Table 2. Study characteristics – use of different forms of cannabis in pain conditions
                                                                                                                                                   PRIMARY
AUTHOR CLINICAL                                                                 INTERVENTION
                 STUDY                                       POPULATION                            DOSING                                          OUTCOME
AND    PAIN                                  DURATION                           (THC POTENCY                 PAIN RELIEF
                 DESIGN                                      (N)                                   FREQUENCY                                       MEASURE (PAIN
DATE   CONDITION                                                                %)
                                                                                                                                                   ASSESSMENT)
SMOKED CANNABIS
                           Randomized,                                                                              Decrease in capsaicin-
           Healthy         double-blind,     4 sessions; 1   N = 19 (M = 11,    Smoked cannabis:                    induced pain by medium
Wallace et
           volunteers      placebo-          week interval   F = 7); 15         low dose (2%),    4 inhalations     dose and increase in
al. (2007)                                                                                                                                      VASPI
           (Capsaicin-     controlled,       between         completed trial    medium dose (4%), per session       capsaicin-induced pain by
(23)
           induced pain)   crossover         sessions        Age: Mean 28.9 y   high dose (8%)                      high dose; no change by low
                           study                                                                                    dose
                           Randomized,       3, 6-hour
                                                                                                                    Decrease in neuropathic
Wilsey et    NP (central   double-blind,     sessions; 3-21 N = 38 (M = 20,     Smoked cannabis:                                                   100-mm VAS; 7-
                                                                                                   9 puffs per 6-   pain; equivalent results for
al. (2008)   and           placebo-          day interval   F = 18);            high dose (7%),                                                    point PGIC scale;
                                                                                                   hour session     high (7%) and low (3.5%)
(24)         peripheral)   controlled,       between        Age: Mean 46 y      low dose (3.5%)                                                    11-point NP scale
                                                                                                                    doses
                           crossover         sessions
                                                                                                                  Decrease in neuropathic pain
                                                                                                                  with high dose (9.4%);
                           Randomized,       Four 14-day
                                                                                                3 inhalations Average daily pain intensity
                           double-blind,     treatment
            Neuropathic                                    N = 23 (M = 11, Smoked cannabis      daily for first 5 less for 9.4% (score 5.4)
                           placebo-          periods (5
Ware et al. pain (post-                                    F = 12); 21      cigarettes weighing days of cycle versus 0% THC (score 6.1) 11-Point Numeric
                           controlled,       days
(2010) (32) traumatic or                                   completed trial  25 mg of four       followed by 9 control (difference = 0.7,       Rating Scale
                           four-period       treatment
            post-surgical)                                 Age: Mean 45.4 y potencies (0%,      day washout       95% CI 0.02 to 1.4)
                           crossover         followed by 9
                                                                            2.5%, 6%, 9.4%)     period
                           study             day washout)
                                                                                                                  Other potencies had modest
                                                                                                                  but nonsignificant decreases
                                                                                                                  Decrease in neuropathic pain
                                                                                                4 inhalations
                           Phase II,                                                                              at maximum tolerable dose
                                                                                                daily for 5
                           randomized,       7 weeks; 5                     Smoked cannabis                       (1-8% THC) of smoked
                                                                                                days after
Ellis et al. HIV           double-blind,     days          N = 28, all male cigarettes ranging                    cannabis
                                                                                                titrating THC                                  DDS; 10 cm VAS
(2009) (27) neuropathy     placebo-          treatment     Age: 48.8 y      between 1 and 8%                      Pain relief (DDS score) and
                                                                                                potency to
                           controlled,       periods                        THC                                   >30% reduction in pain from
                                                                                                tolerance on
                           crossover trial                                                                        baseline: greater with
                                                                                                Day 1
                                                                                                                  cannabis versus placebo
                                                                                                                                                       PRIMARY
AUTHOR CLINICAL                                                                   INTERVENTION
                 STUDY                                         POPULATION                               DOSING                                         OUTCOME
AND    PAIN                                    DURATION                           (THC POTENCY                    PAIN RELIEF
                 DESIGN                                        (N)                                      FREQUENCY                                      MEASURE (PAIN
DATE   CONDITION                                                                  %)
                                                                                                                                                       ASSESSMENT)
                                                                                                                         Decrease in daily pain with
                                                               N = 55: 28                                                smoked cannabis
                             Prospective,                      control, 27                                               >30% reduction in pain from
Abrams et                    randomized,       21 day trial; 5 intervention - 25                     3 inhalations       baseline: 52% of
             HIV                                                                 Smoked cannabis                                                       100 mm VAS;
al. (2007)                   double-blind,     day treatment completed trial                         daily for 5         intervention group; 24% in
             neuropathy                                                          cigarettes weighing                                                   daily diary
(22)                         placebo-          periods         for each category                     days                control group
                                                                                 0.9g (3.56%)
                             controlled                        (M = 48, F = 7);                                          Median reduction on daily
                                                               Age: Mean 50 y                                            VAS: 34% in intervention
                                                                                                                         group; 17% in placebo group
                                                                                                                         Decrease in pain and
                                                                                                                         spasticity with smoked
                                                                                                                         cannabis
                             Randomized,                                                                                                               Ashworth Scale
Corey-                                         17 day trial;                                                             Ashworth scale - smoked
             Multiple        double-blind,                     N = 30 (M = 11,    Smoked cannabis       1 inhalation                                   (spasticity
Bloom et                                       intervention                                                              cannabis modified score by
             sclerosis and   placebo-                          F = 19);           cigarette weighing    daily for 3                                    assessment); VAS
al. (2012)                                     for 3 days                                                                2.74 points more than
             spasticity      controlled,                       Age: Mean 21 y     0.8g (4%)             days                                           (Secondary
(31)                                           total                                                                     placebo
                             crossover trial                                                                                                           Outcome)
                                                                                                                         Pain relief (VAS score) -
                                                                                                                         5.28 points more than
                                                                                                                         placebo
SMOKED CANNABIS - Dronabinol vs Cannabis
                         Randomized,                                                                                     Decrease in experimentally-
              Healthy                                                             Dronabinol (0mg,
                         placebo-      5, 6-hour               N = 30 (M = 15,                          3-7 puffs (70%   induced pain with             McGill Pain
Cooper et individuals                                                             10mg, 20mg) OR
                         controlled,   outpatient              F = 15)                                  of cigarette     dronabinol and cannabis       Questionnaire (post
al. (2013) (experimental                                                          cannabis cigarettes
                         double-       sessions; 2-4           Age: 21-45 y                             pyrolyzed) per   cigarettes; dronabinol        CPT); 100-mm
(26)          ly-induced                                                          (0.00, 1.98, 3.56%
                         dummy,        weeks trial             (Mean 27 y)                              session          produced longer-lasting       VAS
              pain)                                                               THC; ca. 800mg)
                         double-blind                                                                                    decreases
VAPORIZED CANNABIS
                         Randomized, 3, 6-hour
                                                               N = 39 (M = 28,    Vaporized
                         double-blind, sessions; 3 to
                                                               F = 11); current   cannabis: placebo                Decrease in neuropathic
Wilsey et al.            placebo-      14 day                                                       8-12 puffs per                                     100-mm VAS &
               NP                                              or past cannabis   (0%) medium dose                 pain; equivalent results for
(2013) (28)              controlled,   intervals                                                    6-hour session                                     PGIC
                                                               users              (3.53%); low dose                low and medium doses
                         crossover     between
                                                               Age: Mean 50 y     (1.29%)
                         study         sessions
                                                                  Table 2. (Continued)
                                                                                                                                             PRIMARY
AUTHOR CLINICAL                                                           INTERVENTION
                 STUDY                                   POPULATION                            DOSING                                        OUTCOME
AND    PAIN                               DURATION                        (THC POTENCY                   PAIN RELIEF
                 DESIGN                                  (N)                                   FREQUENCY                                     MEASURE (PAIN
DATE   CONDITION                                                          %)
                                                                                                                                             ASSESSMENT)
                                                                         Vaporized                               Decrease in spontaneous
                          Randomized,
                                                                         (aerosolized)         Single dosing     pain scores most with high
Wallace et                placebo-                     N = 16 (M = 9, F
           Diabetic                     4 sessions; 2                    cannabis: low dose    session; 4        dose (7% THC); no              10-cm VAS; foam
al. (2015)                controlled,                  = 7);
           neuropathy                   week intervals                   (1%); medium dose     inhalations per   statistical difference between brush; von Frey
(29)                      crossover                    Age: Mean 56.9 y
                                                                         (4%); high dose       session           medium and low dose (4 and
                          study
                                                                         (7%); placebo (0%)                      1%)
           Chronic pain                                N = 21 (M = 11,                         Inhaled on
                                                                         Vaporized
           (on twice-                                  F = 10);                                evening of
                                                                         cannabis: from
Abrams et daily doses Not placebo-                     Age: Mean 42.9 y                        Day 1, three                                  Drug Effects
                                                                         cigarette 0.9g on                    Decrease in pain ratings on
al. (2011) of sustained controlled, not 5 days total   (morphine cohort)                       times daily on                                Questionnaire
                                                                         average containing                   Day 5 compared to Day 1
(25)       release      randomized                     and 47.1 y                              days 2-4,                                     (subjective)
                                                                         3.56% THC (~96
           morphine or                                 (oxycodone                              morning of
                                                                         mg THC/day)
           oxycodone)                                  cohort)                                 day 5
SMOKED, VAPORIZED, EDIBLE, TOPICAL
                                                                                                                                             Primary: monthly
                                                                                                                                             frequency of
                                                                                                                                             migraine HA
                                                                          Forms of cannabis
                                                                                                                                             Secondary: Type
                                                                          used: vaporized (n
Rhyne et                  Retrospective,                 N = 121 (M = 51,                                        Decrease in monthly         and dose of
             Migraine                                                     = 42), edible (n =
al. (2016)                observational --               F = 63);                              --                frequency of migraine       medical cannabis
             headache                                                     66), topical (n =
(30)                      chart review                   Age = 18-89 y                                           headache                    used, previous and
                                                                          15), smoked (n =
                                                                                                                                             adjunctive
                                                                          55)
                                                                                                                                             migraine therapies,
                                                                                                                                             patient reported
                                                                                                                                             effects
Abbreviations: CPT = cold pressor test; DDS = descriptor differential scale; F = female; M = male; NP = neuropathic pain; PGIC = patient global impression of
   change; THC = delta-9-tetrahydrocannabinol; VAS = visual analog scale; VASPI = visual analog scale of pain intensity; y = years.
               Medical cannabis dosing strategies in pain related conditions                   137
Smoked cannabis
In total, seven studies used smoked cannabis to manage pain associated with various conditions.
Two trials were performed in healthy individuals with no pain history who received a painful
stimulus (23, 26); four studies assessed neuropathic pain (NP) (24, 32), two of which explored
HIV-related neuropathy (22, 27); and one trial assessed MS-associated pain and spasticity (31).
All seven trials demonstrated a significant effect on pain relief compared to controls. The
control in all studies was a placebo cigarette containing 0% THC content, while in one study
smoked cannabis was also compared to an active control (dronabinol, a synthetic cannabinoid).
The dosing frequency and THC content of the intervention drug varied between the studies (see
Table 2).
     Relief of acute pain using smoked cannabis was studied in two trials. The first trial, by
Wallace et al., used 15 healthy volunteers who received intradermal capsaicin-induced
cutaneous pain after four standardized inhalations of smoked cannabis (23). When capsaicin
was used 20 minutes after smoking cannabis (early time course), none of the cannabis potencies
(low [2% THC], medium [4% THC], high [8% THC]), or placebo showed any difference in
pain perception. However, at 55 minutes after smoking cannabis (late time course), capsaicin-
induced pain was decreased by the medium dose (4% THC), but increased by the high dose
(8% THC). Plasma levels of THC and metabolites showed a correlation to the decrease in pain
at the medium dose but this correlation did not exist with the high dose. The authors
hypothesized that there may be another compound not accounted for in the high dose cannabis
that may have contributed to the hyperalgesia. The high dose cannabis was also associated with
increased mild to moderate side effects such as dizziness and somnolence compared to other
doses.
     In another study of cannabis users (≥ 3 cannabis cigarettes at least 4 times per week),
dronabinol was compared to smoked cannabis (26). Pain was experimentally induced using the
Cold Pressor Test, where participants inserted their hands in a warm water bath (37°C) followed
by a cold water bath (4°C) (26). Pain sensitivity (time taken to first report pain sensation), pain
tolerance (time taken to withdraw hand from the cold water) and subjective pain ratings (Pain
Intensity and Bothersomeness Scales) were assessed. As expected, peak effects of the drugs
were seen 15 minutes after cannabis use and 180 minutes after dronabinol use. Compared to
placebo, both smoked cannabis and dronabinol showed decreased pain sensitivity by increasing
the time taken to report the pain sensation (p ≤ 0.01): 3.56% THC (13.1 ± 3.9s from baseline),
20 mg dronabinol (12.1 ± 5.6s from baseline) compared to placebo (0.3 ± 1.0s from baseline).
Pain tolerance was improved (p ≤ 0.05) with low dose cannabis, 1.98% THC and both
dronabinol doses (10mg and 20mg). Although the high dose cannabis (3.56%) increased pain
tolerance in the first hour of administration (9.0 ± 3.0s from baseline), this decreased below
baseline after the first hour and no significant change in tolerance was seen compared to
placebo. In terms of subjective pain ratings of ‘pain intensity’ and ‘bothersomeness’, both
cannabis doses (1.98%, 3.56% THC; p ≤ 0.001) produced a greater decrease compared to high
dose dronabinol (20 mg; p ≤ 0.05). Thus, while smoked cannabis produced more subjective
pain relief than dronabinol, the latter peaked later and produced longer lasting pain relief by
increasing tolerance. Both drugs did not produce any negative subjective effects and cognitive
effects were not assessed.
     There were four studies that looked at relief of neuropathic pain using smoked cannabis.
In all four studies, patients had a mean age range of 45-50 years. They were randomized to
138                     Minhaz Khaiser, Meiko Peng, Michael Lam et al.
smoked cannabis of varying potencies versus placebo (0% THC). Three of the studies had a
crossover design with participant numbers ranging from 23-38 (24, 27, 32) while one had 25
participants in each arm of the study (22). To reduce risk of adverse psychoactive effects in
cannabis naïve patients, two trials limited enrolment to previous cannabis users (22, 24), while
others had no limitations. All studies excluded patients with any psychoactive issues or drug
dependence. Even though neuropathic pain relief was the common outcome, each of the studies
varied in the interventions and pain assessments used.
A 2008 study conducted in central and peripheral neuropathic pain patients by Wilsey et al.
reported an equianalgesic effect between high (7% THC) and low (3.5% THC) potencies of
cannabis cigarettes compared to placebo (24). Pain relief was assessed using the Visual Analog
Scale (VAS) and a 0.0035 reduction per minute in pain intensity was recorded for both doses
(p = 0.016). Another study assessed neuropathic pain caused by trauma or surgery using
three active cannabis potencies (2.5%, 6% and 9.4% THC) smoked for five days (32). An
approximate 11% decrease in daily pain intensity was achieved with the high dose (9.4% THC)
cannabis compared to placebo (p = 0.023). The other potencies (2.5%, 6%) resulted in
nonsignificant, moderate decreases in pain. The higher dose was also associated with more
drowsiness, better sleep with less periods of wakefulness (p < 0.05) there were no reports of
confusion or disorientation which are serious cognitive effects. In the studies assessing HIV
neuropathy, a decrease of > 30% in pain intensity measuring numeral rating scales (clinically
significant decrease) (34) relative to placebo was reported with smoked cannabis use (22, 27).
In both studies, a larger proportion of participants reported reduction in pain intensity when
smoking cannabis (46%-52%) compared to the placebo groups (18%-24%) (22, 27).
Dosing frequency
Three of the studies (HIV and post-traumatic neuropathy) had a range of 3-4 inhalations daily
for five treatment days after which pain relief measures were taken (22, 27, 32). In one study
of mixed neuropathic pain, the intervention had a significantly higher dose, with nine
inhalations over a single 6-hour session (24).
     Some evidence exists for cannabis use in MS-related pain and spasticity. Corey-Bloom et
al. reported reductions in patient reported scores with single inhalation of 4% THC cannabis
for three treatment days (31). There was a decrease in the modified Ashworth scale of 2.74
points representing better spasticity compared to placebo (p < 0.0001). Pain measured using
the VAS decreased by an average of 5.28 points compared to placebo (p = 0.008) (31). Five
participants withdrew from the study due to treatment-related adverse effects in the intervention
arm (feeling euphoria, dizziness, fatigue).
               Medical cannabis dosing strategies in pain related conditions                   139
Vaporized cannabis
Of the three studies using vaporized cannabis, two were conducted to assess neuropathic pain
relief (28, 29). Both were follow-up studies of previous work that had been conducted using
smoked cannabis. In the 2013 follow-up study by Wilsey et al., lower THC concentrations were
used compared to the 2008 study done with smoked cannabis (24, 28). Vaporized cannabis at
a medium (3.53% THC) and low dose (1.29% THC) was inhaled through 8-12 inhalations in a
6-hour session. Even though both doses improved pain tolerance the effect was equianalgesic
when compared to each other. Wallace et al. assessed vaporized cannabis use at three THC
doses (1%, 4%, 7%) in diabetic neuropathy (29). This was a follow up study of one published
in 2007, where similar doses of smoked cannabis were assessed in healthy individuals with
experimentally-induced pain (23). Spontaneous pain scores were significantly decreased by the
high dose (7% THC) compared to other interventions. No statistical difference was reported
between low and medium doses (p = 0.92).
     Vaporized cannabis has also been studied in chronic pain patients currently using opioids.
In one 2011 study, 21 participants with chronic pain on sustained-release morphine (average
dose: 62 mg twice daily) or oxycodone (average dose: 53 mg twice daily) therapy were given
vaporized cannabis (3.56% THC) to augment pain relief (25). Participants in both opioid groups
reported a 27% average decrease in pain measures on Day 5 of treatment compared to Day 1
when they had no exposure to cannabis. Cannabis inhalation was associated to a subjective
“high” that was not seen in opioid use alone.
A recent study in migraine headache (HA) showed a decrease in frequency from 10.4 to 4.6
headaches per month (p < 0.0001) with chronic use of medical cannabis (30). The study was a
retrospective, observational chart review of patients with a follow-up appointment post
migraine diagnosis. In the study, 121 patients used different forms of cannabis, most of whom
used edible (n = 66, 77.4 g monthly), smoked (n = 55, 45.1 monthly), vaporized (n = 42, 74.8
g monthly), and some used topical cannabis (n = 15, 73.4 g monthly). Most patients used
cannabis for both prophylaxis and treatment (90%), though differences in doses have not been
reported. Edible cannabis users were more likely to report negative outcomes including
somnolence, increase headache and difficulty determining dosing and onset of action. While
approximately 85% patients reported reduced frequency of migraine HAs monthly, about 10%
patients who used inhaled forms of cannabis reported complete control of migraine HAs. This
study concluded that cannabis has positive effects in migraine prophylaxis and treatment, and
warrants further prospective studies to assess dose-response relationships in a controlled
environment.
Though the following studies were not included in the final screened trials due to unmet
inclusion criteria (case study; pain relief not assessed), it is worthwhile to explore the evidence
that exists for edible forms of cannabis.
140                     Minhaz Khaiser, Meiko Peng, Michael Lam et al.
Brownie
No randomized trials have been conducted in any pain related indication using cannabis
brownies. A 1988 study by Cone et al. used a double-blind crossover design to assess behavioral
measures for five healthy, male subjects after consumption of cannabis-laced brownies (35).
The participants had a history of cannabis use and the brownies contained 2.8% THC either at
a dose of two 800mg cannabis cigarettes, one 800 mg cigarette or placebo (0% THC) (4, 35).
The intervention group scored higher on behavioral measures assessed using subjective scales
including the Single Dose Questionnaire, Addiction Research Center Inventory (ARCI), and
VAS compared to placebo. However, the peak effects were slow to appear and variable at 2.5-
3.5 hours after brownie consumption unlike smoking cannabis where effects are seen in minutes
(35). Subjects were not restricted in terms of diet that may have contributed to differences in
absorption and inter-subject variability. In another study by Watchel et al., peak plasma THC
levels were analyzed upon consumption of brownies versus smoked cannabis (36). Brownies
containing low dose of THC (9 mg THC/brownie) resulted in a mean peak plasma levels of 5
ng/mL and high dose brownies (13 mg THC/brownie) resulted in mean peak plasma levels of
6-9 ng/mL (whole plant material versus THC) (36). It is interesting to note that the level
associated with euphoria ranges from 50 to 100 mcg/L (37). However, smoking equivalent
THC amounts produced at least five times higher peak plasma THC levels. The dose-dependent
plasma THC levels increased one hour after brownie ingestion, while smoking resulted in a
rapid increase in THC plasma levels (36).
Cannabis oil
No clinical trials were identified for cannabis oil use in pain management. A 1997 case study
of a young male patient with chronic relapsing pain, gastric inflammation and familial
Mediterranean fever symptoms, used cannabis oil as an intervention (38). A peripheral
cannabinoid receptor (unnamed) was identified and localized to the immune system in the
gastrointestinal tract. This investigation hoped to assess the role of cannabinoids in gastric
inflammatory conditions. The patient was randomly assigned to olive or cannabis oil weekly
over six weeks (1 washout week, 1 active week, 2 placebo weeks, 2 active weeks, 1 placebo
week). The dose used was 50 g THC per day as 10 g THC in 5 doses in an effort to minimize
psychotropic effects.(38) The patient continued oral sustained-release morphine (30 g twice
daily) throughout the study and used 10 mg morphine for breakthrough pain management.
During two consecutive placebo weeks, the patient experienced mood disorders due to possible
withdrawal from previous THC use and during the final two weeks he was unable to
differentiate between placebo and drug suggesting possible development of tolerance to the
active medication with two weeks of continuous (38). The most interesting finding in this case
is that the total amount of breakthrough pain medication requirement decreased with use of
cannabis. The patient used 17 tablets of breakthrough morphine during the active weeks versus
41 tablets during placebo weeks. Cannabis oil did not show any anti-inflammatory effects but
this case showed a significant reduction in additional analgesic requirements.
               Medical cannabis dosing strategies in pain related conditions                      141
                                          DISCUSSION
With implementation of new regulations in Canada, eligible individuals with an authorized
prescription can access medical cannabis through licensed producers. A number of Canadians
associate cannabis use with low risk of harm, and it is gaining popularity as a potential treatment
option for unmanageable conditions (39, 40). However, the act of prescribing cannabis is
complicated by the lack of strong evidence on dosing (41). Various professional governing
bodies and researchers have published standards of practice and guidelines to help physicians
navigate through this issue, but further research is warranted to solidify recommendations (20,
42, 43). The College of Family Physicians of Canada recommends physicians follow the
strategy, “Start low, go slow” when prescribing cannabis (37). The recommendation is to
prescribe an inhaled dose of 100-700 mg of no more than 9% THC cannabis daily, titrating to
a maximum 3 g dried cannabis per day. No specific inhalation instructions have been provided
except that patients should start with a single inhalation and appreciate the effects over 4 hours
before increasing (37). One review article made preliminary recommendations to prescribe,
“12g cannabis (9% THC) for 30 days, start with 1 inhalation/day to a maximum of 4
inhalations/day” after conducting a literature review in a chronic non-cancer pain setting (43).
There is a growing body of evidence in support of synthetic THC capsules (dronabinol or
nabilone) and plant extract oromucosal sprays (nabiximols), but less is known about herbal
cannabis (17, 44). The current scoping review aimed to assess existing evidence on dosing and
potency specifically for herbal cannabis in pain related conditions.
Even though nine out of 11 studies included in this review had a randomized, double-blind and
placebo controlled design, common limitations across the studies reduced the quality of
evidence. One issue that was recognized by most studies is the small participant size ranging
from 16-27 participants in the intervention groups. With the exception of one study, where HIV
neuropathy patients were all male, there is a fairly good representation of both genders in other
trials (27). Thus, the generalizability of the data is limited. The crossover design of most studies
makes blinding of cannabis versus placebo challenging. The psychoactive effect of THC
(especially at high potencies) contributes in patients correctly guessing the intervention they
receive. Ellis et al. tried to correct for this factor by asking participants to report what they think
they received at several points during the study (27). In the first treatment week, guessing was
no better than chance and cannabis was still shown to be better than the control group in
providing pain relief. In other studies, treatment effects could have been enhanced due to
unmasking of interventions. However, using more than one intervention group, for example
2.5% versus 6% versus 9.4% THC would increase the likelihood of blinding among
intervention groups as participant may not be able to determine the exact potency (32).
142                      Minhaz Khaiser, Meiko Peng, Michael Lam et al.
This review mainly assessed herbal cannabis delivered through inhalation (smoking or
vaporizing) and commentary has been included on cannabis brownies and oil. Evidence shows
that oral administration has a slower onset, longer duration of action, and lower peak levels of
medication in the blood when compared to inhalation (8). Peak plasma THC levels can be at
least 5 times higher through smoking compared to oral routes (36). Even though some
behavioral changes were noticed using 2.8% THC cannabis-laced brownies, these appeared
2.5-3.5 h after consumption (35). Thus, the typical side effect of THC, euphoria, and other
positive reinforcement effects such as relaxation and enhanced sensory experiences occurs
rapidly through inhaled forms (~15 minutes) compared to oral administration (45). Individual
physiological differences such as rate of gastrointestinal absorption, degree of first-pass
metabolism and genetic makeup of the participant may also contribute to inter-subject
variability with oral cannabis. Since inhalation provides a direct and fast onset of pain relief, it
is the more preferred route of administration. However, there is a proven relationship for THC
dose-related adverse neurologic or psychoactive effects (e.g., dysphoria, sedation and poor
concentration) with inhaled cannabis (16). In one study, the ratings for “feeling high” (p <
0.003) and “feeling stoned” (p < 0.004) were highest with the higher dose (3.53% THC) versus
the lower dose (1.29% THC) (28). Since oral cannabis results in lower plasma levels of THC
these may cause less psychoactive effects and may be designed to reduce the euphoric “high”
that is experienced by inhaled cannabis. This is relevant for certain patient populations (e.g.,
older, work requiring high cognitive performance) who may opt for oral cannabis to avoid the
“high” even if it means a delayed pain relief effect. Low THC potencies used in low doses have
also shown to cause cognitive impairment in some patients that lasts up to 24 hours (4). Thus,
even if oral cannabis or low THC potencies of inhaled cannabis are selected to prevent
psychoactive effects, there is no guarantee of how a patient may respond.
Smoking is the act of burning the plant material and inhaling the smoke while vaporizing is
heating the herb below point of combustion (7). Taking into account the hazards of smoking,
investigators of two follow-up studies chose to repeat their initial smoking studies using
vaporized cannabis as this would reduce exposure to carcinogens (23, 24, 28, 29). Smoking is
not an optimal delivery system as similar carcinogenic materials usually found in tobacco
smoke are also present in cannabis smoke (9). Vaporizing cannabis can produce carbon
monoxide as a by-product but it is comparably in much smaller amounts compared to smoking
(7). Thus, in agreement with the recommendations by governing bodies, vaporizing should be
the chosen mode of delivery (20, 43).
In most studies, the “Foltin Puff Procedure” was used which standardized the inhalation
protocol. Participants mostly self-administered cannabis while being given verbal cues, but in
one study were taught the inhalation method to be used at home (32). Participants in this study
               Medical cannabis dosing strategies in pain related conditions                    143
collected daily urine samples and returned all capsules (containing herbal cannabis to be used
in a titanium pipe) at the end of each intervention period. Investigators concluded compliance
was excellent as all dispensed capsules were returned, and urine and plasma THC assays were
consistent with results (32). Participants in a clinical trial are under supervision and have regular
follow-ups in a controlled environment, but this is not the case in clinical practice. Practitioners
who choose to prescribe cannabis need to consider various methods to prevent or control
substance abuse, such as prescribing small amounts at a time, or ordering regular urine testing
to reduce risk of diversion or abuse.
Dose-response relationship
Trials studying smoked cannabis for neuropathic and MS pain conditions had the shortest
intervention periods, lasting 1-5 days. The dosing frequency ranged from one inhalation daily
to nine inhalations in a 6-hour session, with majority of the studies ranging from 3-4 inhalations
per day. A number of the studies defined low, medium, or high dose using THC percentage by
weight.
     The wide variation in the design of the trials makes it challenging to compare them head-
to-head but some important conclusions can be highlighted. Overall, use of cannabis reduced
participant reported pain measures compared to placebo. This was observed in studies using a
single potency of THC (~4%) in HIV related neuropathy and MS related pain settings (22, 31)
     Similar studies of smoked and vaporized cannabis in neuropathic pain settings found
equianalgesic effects between low, medium, and high THC potencies (24, 28). In other studies,
decrease in spontaneous pain scores was most with the highest dose studied (25, 28). These
results highlight the challenge that exists in selecting the best initial cannabis potency for
treatment. One study avoided this dilemma by first titrating participants to a maximum
tolerance ranging 1-8%, and then performed the intervention phase of the trial, which resulted
in successful pain reduction from baseline (27). Therefore, titration by “starting low, going
slow” is the best dosing strategy as 1) lower potencies have shown equivalent efficacy to higher
doses; 2) all patients may not equally tolerate higher potencies of cannabis.
Dosing recommendation
In neuropathic pain settings, controlled trials have shown efficacy of THC doses from 1% to
9%. The best dosing strategy in this group is to start at the lower THC doses based on patient
history of cannabis use (e.g., 1-2% THC) up to 3-4 vaporized inhalations daily (separated by
45 seconds based on Foltin procedure) (33). For cannabis-naïve patients, it should be required
for patients to start at the lowest THC dose (i.e., 1% THC) to avoid undesirable side effects and
titrate up to pain relief (19). Cannabis-naïve patients should be counseled to separate inhalations
by at least 30 minutes to assess side effect and prevent overdosing. For non-neuropathic pain
the same recommendations may be followed as above. Cannabis oil may be dosed using
dronabinol/Marinol® (oral capsules of synthetic THC in sesame oil) as a reference as no
clinical trials exist (19). Average dose of Marinol® used is 20 mg THC per day and doses range
from 2.5 mg to maximum 210 mg THC per day. Just as with the inhaled cannabis, oils should
be prescribed at the lowest dose (e.g., 2.5 mg THC) and then titrated to effect (19). Patients
144                       Minhaz Khaiser, Meiko Peng, Michael Lam et al.
should wait at least two hours between administrations of single oral doses as acute effects may
last up to 4 hours (4). To start, dried cannabis should be prescribed at 1 g per day (30 g/month)
to a maximum of 3 g per day (90 g/month) (4). The amount prescribed should follow the MMPR
regulation: “30 times the daily quantity of dried marijuana indicated by your healthcare
practitioner on your medical document, or 150 grams of dried marijuana, whichever is less”
(18).
Due to the nature of this scoping review where MeSH terms were used in databases deemed
most appropriate, it is not possible to ensure all relevant studies were gathered for inclusion.
Cannabis dosing in pain settings has very limited evidence and clinical studies of cannabis-
based edible products (e.g., oils, teas, brownies etc.) are non-existent. Case studies and animal
studies were excluded and should be considered for inclusion moving forward as they may
provide some perspective on novel dosing strategies that may be of clinical importance.
                                          CONCLUSION
Even though medical cannabis is not approved as a therapeutic agent in Canada, it is available
for medical use by patients with an authorized prescription. Current literature on herbal
cannabis mostly involves smoked and vaporized mode of delivery, and very little evidence
exists regarding oral forms such as brownies and oils. Smoking is not an optimal delivery
system due to toxic by-products and thus, vaporizing cannabis is preferred. The studies
identified in this review are limited by large variability in dosing strategies, small sample sizes
and short intervention periods. Standardized studies regarding orally consumed cannabis (oils,
brownies etc.) are essentially non-existent and limit our ability to make substantial conclusions.
It is best to choose a low potency to start treatment with and titrate to an optimal dose that
provides pain relief, improves function with minimal adverse effects.
                                          REFERENCES
[1]   Borgelt LM, Franson KL, Nussbaum AM, Wang GS. The pharmacologic and clinical effects of medical
      cannabis. Pharmacotherapy 2013;33(2):195–209.
[2]   Poznyak V. Global epidemiology of cannabis use and implications for public health. Alcohol Alcohol
      2014;49(1):1–69.
[3]   Elsohly M, Slade D. Chemical constituents of marijuana: The complex mixture of natural cannabinoids.
      Life Sci 2005;78(5):539–48.
[4]   Abramovici H. Information for health care professionals: Cannabis (marijuana) and the cannabinoids.
      Health Canada 2013. URL: http://www.hc-sc.gc.ca/dhp-mps/marihuana/med/infoprof-eng.php.
[5]   Ashton HC. Pharmacology and effects of cannabis: a brief review. Br J Psychiatry 178(2):101–6.
[6]   Hazekamp A, Ware MA, Muller-Vahl K, Abrams D, Grotenhermen F. The medicinal use of cannabis
      and cannabinoids-an international cross-sectional survey on administration forms. J Psychoact Drugs
      2013;45(3):199–210.
                Medical cannabis dosing strategies in pain related conditions                              145
[7]    Grant I, Atkinson JH, Gouaux B, Wilsey B. Medical marijuana: Clearing away the smoke. Open Neurol
       J 2012;6(1):18–25.
[8]    Huestis M. Human cannabinoid pharmacokinetics. Chem Biodivers 2007;4(63):1770–804.
[9]    Gieringer DH. Cannabis “vaporization”: A promising strategy for smoke harm reduction. J Cannabis
       Ther 2001;1(3-4):153–70.
[10]   Abrams D, Vizoso H, Shade S, Jay C, Kelly M, Benowitz N. Vaporization as a smokeless cannabis
       delivery system: A pilot study. Clin Pharmacol Ther 2007;82(5):572–8.
[11]   Fischer B, Imtiaz S, Rudzinski K, Rehm J. Crude estimates of cannabis-attributable mortality and
       morbidity in Canada-implications for public health focused intervention priorities. J Public Health (Oxf)
       2016;38(1):183–8.
[12]   Hama AT, Sagen J. Cannabinoid receptor-mediated antinociception with acetaminophen drug
       combinations in rats with neuropathic spinal cord injury pain. Neuropharmacology 2010;58(4-5):758–
       66.
[13]   Aggarwal S, Carter G, Sullivan M, ZumBrunnen C, Morrill R, Mayer J. Medicinal use of cannabis in
       the united states: historical perspectives, current trends, and future directions. J Opioid Manag
       2009;5:153–68.
[14]   Jensen B, Chen J, Furnish T, Wallace M. Medical marijuana and chronic pain: A review of basic science
       and clinical evidence. Curr Pain Headache Rep 2015;19(10):1–9.
[15]   Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez A V, et al. Cannabinoids for
       medical use: A systematic review and meta-analysis. JAMA 2015;313(24):2456.
[16]   Deshpande A, Mailis-Gagnon A, Zoheiry N, Lakha S. Efficacy and adverse effects of medical marijuana
       for chronic noncancer pain. Can Fam Physician 2015;61:372–81.
[17]   Lynch ME, Ware MA. Cannabinoids for the treatment of chronic non-cancer pain: An updated
       systematic review of randomized controlled trials. J Neuroimmune Pharmacol 2015;293–301.
[18]   Government of Canada. Marihuana for Medical Purposes Regulations, SOR/2013-119. Government of
       Canada Department of Justice 2015. URL: http://www.laws-lois.justice.gc.ca/eng/regulations/SOR-
       2013-119/.
[19]   Health Canada. Authorized Licensed Producers under the Marihuana for Medical Purposes
       Regulations. Health Canada 2016. URL: http://www.hc-sc.gc.ca/dhp-mps/marihuana/med/daily-
       quotidienne-eng.php.
[20]   College of Physicians and Surgeons of Ontario. Marijuana for medical purposes. CPSO 2014. URL:
       http://www.cpso.on.ca/CPSO/media/documents/Policies/Policy-Items/Marijuana-for-Medical-
       Purposes.pdf?ext=.pdf.
[21]   Arksey H, O’Malley L. Scoping studies: Towards a methodological framework. Int J Soc Res Methodol
       2005;8:19–32.
[22]   Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful HIV-associated
       sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007;68(7):515–21.
[23]   Wallace M, Schulteis G, Atkinson JH, Wolfson T, Lazzaretto D, Bentley H, et al. Dose-dependent
       effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers.
       Anesthesiology 2007;107(5):785–96.
[24]   Wilsey B, Marcotte T, Tsodikov A, Millman J, Bentley H, Gouaux B, et al. A randomized, placebo-
       controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain 2008;9(6):506–21.
[25]   Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL. Cannabinoid-opioid interaction in chronic
       pain. Clin Pharmacol Ther 2011;90(6):844–51.
[26]   Cooper ZD, Comer SD, Haney M. Comparison of the analgesic effects of dronabinol and smoked
       marijuana in daily marijuana smokers. Neuropsychopharmacology 2013;38(10):1984–92.
[27]   Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, et al. Smoked medicinal
       cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology
       2009;34(3):672–80.
[28]   Wilsey B. Low dose vaporized cannabis significantly improves neuropathic pain. J Pain
       2013;14(2):136–48.
[29]   Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful
       diabetic neuropathy. J Pain 2015;16(7):616–27.
146                         Minhaz Khaiser, Meiko Peng, Michael Lam et al.
[30]   Rhyne DN, Anderson SL, Gedde M, Borgelt LM. Effects of medical marijuana on migraine headache
       frequency in an adult population. Pharmacotherapy 2016;36(5):505–10.
[31]   Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H, et al. Smoked cannabis for
       spasticity in multiple sclerosis: a randomized, placebo-controlled trial. CMAJ 2012;184(10):1143–50.
[32]   Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, et al. Smoked cannabis for chronic
       neuropathic pain: A randomized controlled trial. CMAJ 2010;182(14):e694-701.
[33]   Chait LD, Corwin RL, Johanson CE. A cumulative dosing procedure for administering marijuana smoke
       to humans. Pharmacol Biochem Behav 1988;29(3):553–7.
[34]   Farrar JT, Young JP, LaMoreaux L, Werth JL, Poole RM. Clinical importance of changes in chronic
       pain intensity measured on an 11-point numerical pain rating scale. Pain 2001;94(2):149–58.
[35]   Cone EJ, Johnson RE, Paul BD, Mell LD, Mitchell J. Marijuana-laced brownies: Behavioral effects,
       physiologic effects, and urinalysis in humans following ingestion. J Anal Toxicol 1988;12(4):169–75.
[36]   Wachtel S, ElSohly M, Ross S, Ambre J, De Wit H. Comparison of the subjective effects of delta-9-
       tetrahydrocannabinol and marijuana in humans. Psychopharmacology (Berl) 2002;161(4):331–9.
[37]   College of Family Physicians of Canada. Authorizing Dried Cannabis for Chronic Pain or Anxiety:
       Preliminary Guidance from the College of Family Physicians of Canada. Mississauga, ON: College of
       Family Physicians of Canada, 2014.
[38]   Holdcroft A, Smith M, Smith B, Hodgson H, Evans FJ. Clinical trial experience with cannabinoids.
       Pharm Sci 1997;3(11):546–50.
[39]   Poulin C, Elliott D. Alcohol, tobacco and cannabis use among nova scotia adolescents: Implications for
       prevention and harm reduction. CMAJ 1997;156(10):1387–93.
[40]   Duff C, Asbridge M, Brochu S, Cousineau M-M, Hathaway AD, Marsh D, et al. A canadian perspective
       on cannabis normalization among adults. Addict Res Theory 2012;20(4):271–83.
[41]   Abrams DI. Integrating cannabis into clinical cancer care. Curr Oncol 2016;23:S8–14.
[42]   College of Physicians and Surgeons of British Columbia. Professional Standards and Guidelines.
       Vancouver, British Columbia: College of Physicians Surgeons of British Columbia, 2015.
[43]   Kahan M, Srivastava A, Spithoff S, Bromley L. Prescribing smoked cannabis for chronic noncancer
       pain: preliminary recommendations. Can Fam Physician 2014;60(12):1083–90.
[44]   Martin-Sanchez E, Furukawa TA, Taylor J, Martin JLR. Systematic review and meta-analysis of
       cannabis treatment for chronic pain. Pain Med 2009;10(8):1353–68.
[45]   Ashton CH. Pharmacology and effects of cannabis: A brief review. Br J Psychiatry 2001;178:101-6.
In: Cannabis: Medical Aspects                                              ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                     © 2017 Nova Science Publishers, Inc.
Chapter 12
                                 Stephanie Stockburger*, MD
    Division of Adolescent Medicine, UK Healthcare, Department of Pediatrics, Lexington,
                                      Kentucky, US
       The legalization of marijuana for medical use is increasingly common in the United States.
       The marijuana plant contains a number of cannabinoids along with chemical delta-9-
       tetrahydrocannabinol (THC) which is known to be psychoactive. In states where medical
       marijuana is legal, physicians may be trained to certify patients for medical marijuana use.
       Patients then take their certification and obtain medical marijuana from a dispensary. The
       dispensary provides advice on which marijuana species or strain to purchase as well as
       dosing and administration. Forms of medical marijuana are the same as recreational
       marijuana. Forms include dried leaves or buds for smoking or ingestion and hashish which
       may also be smoked or ingested. Marijuana concentrates are becoming more popular as
       they are easier to conceal and require a smaller amount to produce the desired effect. Types
       of concentrates include butane honey oil, CO2 honey oil, wax or budder, and shatter.
       Dabbing is a technique used for consuming concentrates that involves placing a “dab” of
       the concentrate on a heated surface and inhaling the vapors. As products are not regulated
       by the United States Food and Drug Administration, there is concern that potency and
       product-labeling of THC content varies widely among dispensaries and patients are at risk
       for under- and over-dosing. Overall, more research is needed about the effects of specific
       cannabinoids on individual body systems and health conditions in order to standardize
       forms of administration and their efficacy as well as dosing.
                                            INTRODUCTION
The legalization of marijuana for medical use is increasing in the United States. Marijuana is
the dried leaves, flowers, stems, and seeds from the hemp plant, Cannabis sativa (1, 2).
Marijuana contains numerous cannabinoids. The chemical delta-9-tetrahydrocannabinol (THC)
is a known mind-altering chemical that is found within the plant. Currently, there are two
*
    Correspondence: Stephanie J Stockburger, MD, Division of Adolescent Medicine, UK Healthcare, Department of
       Pediatrics, KY Clinic Room J413, Lexington, KY 40536-0284, United States. E-mail: sjstoc2@uky.edu.
148                                    Stephanie Stockburger
United States Food and Drug Administration (FDA) approved cannabinoids: dronabinol and
nabilone. The FDA-approved indications for these drugs are nausea and vomiting associated
with chemotherapy and appetite stimulation in acquired immunodeficiency syndrome (AIDS)
(1).
     Marijuana (as opposed to the isolated cannabinoids mentioned above) is classified as a
schedule I drug which means it has no currently accepted medical use and has a high potential
for abuse (1). However, our current law permits individual states to allow physicians to certify
marijuana use for certain medical conditions (3). Many states agree that Alzheimer’s disease,
amyotrophic lateral sclerosis, cachexia/wasting syndrome, cancer, Crohn’s disease, epilepsy
and seizures, glaucoma, hepatitis C virus, human immunodeficiency virus/acquired
immunodeficiency syndrome, multiple sclerosis and muscle spasticity, severe and chronic pain,
and severe nausea are conditions that physicians can certify patients for medical marijuana use
(3). Typically, it is recommended that the FDA-approved cannabinoids dronabinol and
nabilone are tried initially. Then if patient has minimal or no improvement, therapy may be
escalated to medical marijuana use that is certified by the physician. The patient then goes to a
dispensary and receives advice on which marijuana species or strain to purchase as well as
dosing and administration (1). The forms and administration of medical marijuana are identical
to the use of recreational marijuana.
                          FDA-APPROVED CANNABINOIDS
Currently, there are two isolated cannabinoids that are approved by the FDA. These are
dronabinol and nabilone. They are approved for the treatment of chemotherapy-induced nausea
and vomiting as well as appetite stimulation in patients with acquired immunodeficiency
syndrome (AIDS) (4). Both drugs are available in capsule form. Dosing of dronabinol is 2.5
mg to 10 mg three or four times daily. Dronabinol is available in a 2.5 mg capsule, 5 mg capsule,
and a 10 mg capsule (4). It is considered a controlled substance, category C-III. For appetite
stimulation, the initial recommended dose is 2.5 mg twice daily (before lunch and dinner) or
once daily if dose is not tolerated (4). Dose can be titrated up based on response and tolerability
to a maximum of 20 mg per day (4). For chemotherapy-induced nausea and vomiting the
recommended dose is 5 mg/m2 administered 1 to 3 hours before chemotherapy, then give
5mg/m2/dose every 2 to 4 hours after chemotherapy for a total of 4 to 6 doses/day (4). Dose
can be increased in increments of 2.5 mg/m2 based on response and tolerability with a
maximum dose of 15 mg/m2/dose. Off-label dosing of chemotherapy-induced nausea and
vomiting that is refractory is 2.5 to 10 mg 3 or 4 times daily (4).
     Nabilone is FDA approved for treatment of nausea and vomiting associated with cancer
chemotherapy (5). It also comes in a capsule form (5). Capsules are 1 mg. Dosing instructions
are 1-2 mg twice daily with a maximum of 6 mg in 3 doses daily (5). It is recommended to
begin with the lower dose and increase if needed. Nabilone is also considered a controlled
substance and is category C-II (5). It is approved for treatment for adults but use in pediatric
patients is considered off-label (5).
                        How to administrate cannabis and efficacy                              149
                                 MEDICAL MARIJUANA
As mentioned previously, the use of medical marijuana can be certified by a physician for
certain conditions within state laws. States that allow physicians to certify medical marijuana
have their own list of approved conditions that may be managed with medical marijuana (1).
Each state that has legalized medical marijuana also has a legal limit which may be something
like “1-2 ounces every 30 days”. The legal limit varies widely by state (1). It is important to
note that physicians are not prescribing marijuana but instead are certifying that the patient has
a medical condition that may benefit from medical marijuana use and that the physician has
discussed the risks and benefits (1). Certification must state the medical condition that the
physician believes would be treated effectively with the medical marijuana (1). In some states,
the recommended amount of marijuana needed to treat the condition must also be stated on the
certification (1). Marijuana cannot be prescribed due to its status at the federal level of being a
schedule I drug which makes it illegal (1). It is not regulated by the FDA or dispensed by
pharmacies (1). Physicians must have training in prescribing medical marijuana which usually
consists of continuing medical education (CME) activities prior to certifying patients for
medical marijuana use (1).
     When physicians are treating patients for conditions that would otherwise be treated by
marijuana itself, it is reasonable to start therapy with dronabinol or nabilone (1). If these
medications are not successful, treatment may be escalated to marijuana itself (1). Marijuana
contains a number of cannabinoids and it is currently not known how individual cannabinoids
affect the various diseases that are currently managed with medical marijuana (1). Conditions
which are permissible for treatment with medical marijuana vary by state. However, there are
a number of conditions that states agree upon in which management with medical marijuana is
permissible (3). These conditions include Alzheimer’s disease, amyotrophic lateral sclerosis
(ALS), cachexia/wasting syndrome, cancer, Crohn’s disease, epilepsy and seizures, glaucoma,
hepatitis C virus, HIV/AIDS, multiple sclerosis and muscle spasticity, severe and chronic pain,
and severe nausea (3). Once a patient begins medical marijuana treatment, close physician
follow-up is very important as it is with any medication that could have significant adverse
effects and abuse potential (1). It is recommended that the patient be seen for follow-up within
30 days with additional telephone contact as necessary (1). After this, the patient may be
followed monthly for three months and then in intervals depending on the clinical situation (1).
     Medical marijuana is administered in the same way as recreational users use the drug.
When a patient receives certification from a physician for treatment with medical marijuana,
they take the certification to a dispensary (1). The dispensary provides advice on which
marijuana species or strain to purchase as well as dosing and administration (1). Forms of
marijuana include dried leaves and buds, resin from the flowers, and extracts. Only the leaves
and buds of the female Cannabis Sativa and Cannabis indica plants contain psychoactive
properties (6). The male plants do not contain THC and therefore do not have psychoactive
properties (6).
                                          SMOKING
Smoking leaves and buds of the marijuana plant is a common way to use marijuana
recreationally or medically. Joints (also called “spliffs or doobies” are hand-rolled cigarettes
150                                    Stephanie Stockburger
that contain marijuana leaves and buds (2). A blunt is an emptied cigar that is partly or
completely refilled with marijuana (6). The lumpy texture of marijuana as well as the distinct
odor of marijuana distinguishes it from tobacco (6). The marijuana may be sprayed with
contaminants such as other psychoactive drugs to enhance effects of poor quality, stale, or male
(not psychoactive) marijuana plants (6). Pipes and water pipes may also be used to smoke
marijuana.
    Hashish (hash, for short) is made by shaking the buds of marijuana and dropping the resin
glands onto silk screens (6, 7). The resin glands are then sieved through the silk screens to
create kief. Kief has a loose powdery consistency. Kief is normally compressed to form a block
of hash. Kief and hash may be smoked in pipes, water pipes, bongs or mixed with tobacco and
smoked (6). The different names for hashish include black, goldseal black, redseal black, and
Morrocan (Rocky for short) (6). Hashish is an oily, solid substance (6).
                                           EDIBLES
Marijuana can be mixed in food such as brownies, cookies or candy (2). It can also be brewed
as a tea (2). It can also be used like a kitchen herb and added to many types of food (6). Hashish
and hashish oil can also be dissolved into milk and consumed in drinks (6). Cannabis is an oil-
based substance and the emulsive properties of milk allow it to be dissolved. When marijuana
is ingested, compared to inhaling the smoke or vapors of marijuana, it has a delayed-onset of
effect, and tends to last for a longer period of time (8). It is estimated that 16% to 26% of
patients using medical cannabis consume edible products (9). Oral consumption does not have
the harmful effects of smoking but users may have a more difficult time titrating their dose to
desired effect (10).
                                     CONCENTRATES
Concentrates contain a higher percentage of THC than marijuana leaves or hash products (7).
All concentrates must be made by using some type of solvent to extract the THC (7). Toward
the end of the concentrate-making process, the solvents and plant matter are removed (7).
Solvents used are either butane or carbon dioxide (CO2) (7). There is concern that it is difficult
to completely remove the solvent from the concentrate. Health concerns exist about consuming
concentrate that still contains butane (7). However, professional concentrate manufacturers
have supposedly become extremely efficient at removing butane (7). In addition, because the
process usually involves butane, which is lighter fluid, individuals who have attempted to create
extracts at home have caused fires and explosions and have been seriously burned and injured
(2).
     Concentrates may be in the form of oil, wax, or shatter. Hash oil or honey oil is a gooey
liquid that is usually used in vape pens (7). As above, the extraction process either involves the
use of butane (butane honey oil or BHO) or CO2 (CO2 honey oil). Wax or budder is a soft solid
that has a texture like lip balm (2). It is created by whipping hash oil during the purging process
and has a consistency that is comparable to earwax (7). The percentage of THC between oil
and wax are similar (7). Shatter is a hard, amber-colored solid that is a refined version of butane
                        How to administrate cannabis and efficacy                           151
honey oil (2, 7). It takes multiple steps to make in order to extract all the plant matter and
solvents and is usually made in a pressure vacuum (7). It is usually a thin cake, is yellow or
amber in color, and “shatters” when you break a piece off (7). Shatter is the most potent
concentrate and can contain 90% THC (7).
                                         DABBING
Dabbing is a technique that is used for consuming concentrates (7). It involves placing a “dab”
of concentrate onto a heated surface which vaporizes the concentrate. The concentrate is then
inhaled (7). There is a recent increase in “dabbing” among recreational marijuana users as well
as medical marijuana users (11). Users are recommended to start with a single inhalation of
marijuana vapor and monitor for effect (1). If twenty minutes pass without effect, the patient
may take two inhalations and monitor for another twenty minutes (1).
products from three major metropolitan areas (10). Dispensaries were located by an internet
directory and three dispensaries from each of the three major metropolitan areas were randomly
identified (10). Individuals with physician certification for medical cannabis use purchased
products from the dispensaries. The samples were tested with high-performance liquid
chromatography. Researchers noted that the products failed to meet basic label accuracy
standards for pharmaceuticals (10). They found that greater than 50% of products evaluated
had significantly less cannabinoid content than labeled, with some products containing
negligible amounts of THC (10). They noted that other products contained significantly more
THC than labeled. If products are over-labeled, the medical benefits may be negligible (10). If
products are under-labeled, the higher THC content puts patients at risk of experiencing
increased adverse effects (10).
                                           DOSING
Dosing for the FDA-approved products dronabinol and nabilone is well-established. However,
the dosing for medical marijuana is not standardized. According to the World Health
Organization, a standard marijuana cigarette contains as little as 0.5 g of marijuana (1). As an
example, in Massachusetts, state law allows a 60-day supply of 10 oz of medical marijuana (1).
A 60-day supply of 10 oz is up to 560 marijuana cigarettes or about 10 per day (1). If a patient
smokes 1 to 2 marijuana cigarettes a day, they would need about 0.5 to 1 oz of marijuana per
month (1).
     It is estimated that ingesting marijuana requires about three to five times the smoked dosage
(8). When cannabis is ingested, the effects are spread out over a longer period of time (8). This
may be useful for patients who are having trouble with sleep or situations where smoking is
impractical or impossible (8). However, ingested cannabis can be harder to titrate due to its
delayed onset and an individual’s metabolic activity (8).
     It is recommended that medical marijuana users have an accurate scale to weight, measure,
track and titrate their dosage and supply of cannabis (8). Patients tend to “stockpile” their
marijuana as they cannot purchase it at the pharmacy (8). One author recommends a personal
supply of three to six pounds (8). There is concern that potency diminishes a little with time
but cannabis can be reportedly stored in a cool, dark, dry place for years without significant
loss of effect (8).
                                       CONCLUSION
In conclusion, there are a number of different forms of administration of medical marijuana. At
this time, medical marijuana is not regulated by the FDA and therefore dispensaries often have
under- or over-labeled THC content of their products. Marijuana contains a number of different
cannabinoids which may have different effects on different medical conditions. There may be
a role for the non-psychoactive CBD oil in treatment for a number of medical conditions. More
research is needed to safely recommend marijuana forms and route of administration as well as
specific strains and species of the plant.
                          How to administrate cannabis and efficacy                                    153
                                           REFERENCES
[1]    Hill KP. Medical marijuana for treatment of chronic pain and other medical and psychiatric problems.
       JAMA 2015;313(24):2474-83.
[2]    National Institute on Drug Abuse. DrugFacts: Marijuana. URL: https://www.drugabuse.gov/
       publications/drugfacts/marijuana.
[3]    Belendiuk KA, Baldini LL, Bonn-Miller MO. Narrative review of the safety and efficacy of marijuana
       for the treatment of commonly state-approved medical and psychiatric disorders. Addict Sci Clin Pract
       2015;10:1-10.
[4]    UpToDate. Dronabinol: Drug information. URL: http://www.uptodate.com/contents/dronabinol-drug-
       information?source=search_result&search=dronabinol&selectedTitle=1%7E25.
[5]    UpToDate. Nabilone: Drug information. URL: http://www.uptodate.com/?source=search_
       result&search=nabilone&selectedTitle=1%7E13.
[6]    Hartney E. Types of marijuana: How to recognize the many different forms of marijuana. URL:
       https://www.verywell.com/types-of-marijuana-22323.
[7]    PotGuide.com Colorado. Different types of marijuana concentrates available in Colorado. URL:
       https://www.coloradopotguide.com/colorado-marijuana-blog/2014/february/06/different-types-of-
       marijuana-concentrates-available-in-colorado/
[8]    Conrad C. Cannabis yields and dosage. URL: http://www.safeaccessnow.net/yieldsdosage.htm.
[9]    Grella CE, Rodriguez L, Kim T. Patterns of medical marijuana use among individuals sampled from
       medical marijuana dispensaries in Los Angeles. J Psychoactive Drugs 2014;46(4):267-75.
[10]   Vandrey R, Raber JC, Raber ME, Douglass B, Miller C, Bonn-Miller MO. Cannabinoid dose and label
       accuracy in edible medical cannabis products. JAMA 2015;313(24):2491-3.
[11]   Stogner JM, Miller BL. Assessing the dangers of “dabbing”: mere marijuana or harmful new trend?
       Pediatrics 2015;136(1):1-3.
[12]   Leaf science. 5 must-know facts about cannabidiol (CBD). URL: http://www. leafscience.com/
       2014/02/23/5-must-know-facts-cannabidiol-cbd/
[13]   Mehmedic Z, Chandra S, Slade D, Denham H, Foster S, Patel A, et al. Potency trends of Δ9-THC and
       other cannabinoids in confiscated cannabis preparations from 1993 to 2008. J Forensic Sci 2010;55
       (5):1209-17.
[14]   Wright S, Yadav V, Bever C Jr, Bowen J, Bowling A, Weinstock-Guttman B, et al. Summary of
       evidence-based guideline: complementary and alternative medicine in multiple sclerosis: report of the
       Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2014;83
       (16):1484-6.
In: Cannabis: Medical Aspects                                                           ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                                  © 2017 Nova Science Publishers, Inc.
Chapter 13
             Cannabis and cannabis derivatives are sometimes used to relieve pain. Objective: To
        conduct a scoping review to explore the extent of the literature on the efficacy, safety, and
        side effects of cannabis and cannabis derivatives as a treatment for pain. Methods: The
        English-language literature was searched using electronic databases for studies published
        from 1960 to August 15, 2016. All randomized controlled trials that compared cannabis to
        a control and that examined pain as an outcome were included. Data on demographic and
        clinical characteristics, study duration, intervention duration, and outcomes were
        abstracted. Results: Of 4,472 articles identified through the literature search, only 28
        studies satisfied eligibility criteria. An additional five were identified through hand search
        Most studies had very small sample sizes. The primary methods of administration were
        oromucosal spray of nabiximol, oral ingestion of cannabis extract capsules, and inhalation
        of smoked cannabis. Overall, nabiximol oromucosal sprays resulted in reductions in pain
        that were statistically significant but of variable clinical relevance. Studies of oral cannabis
        extracts nabilone and dronabinol yielded mixed results, with some studies demonstrating
        effectiveness and others being negative. Studies of smoked cannabis consistently
        demonstrated statistically significant and clinically relevant reductions in neuropathic pain.
        Conclusions: Published research on the efficacy of cannabis as a treatment for pain is
        extremely limited. Evidence of effectiveness was strongest for smoked cannabis for
        neuropathic pain.
                                                    INTRODUCTION
Cannabis and cannabis derivatives have been used since ancient times as an analgesic to relieve
pain from a variety of conditions. Although long considered a natural remedy for
*   Corresponding author: Hance Clarke, MD, PhD, Staff Anesthesiologist, Director of Clinical Pain Services and Medical Director of
        Pain Research Unit, Toronto General Hospital, 200 Elizabeth Street, Toronto, ON Canada. E-mail: Hance.Clarke@uhn.ca.
156                            Jonathan K Hwang and Hance Clarke
various conditions, the evidence for its use has not been systematically reviewed until relatively
recently. This scoping review will explore the extent of the literature on the efficacy, safety,
and side effects of cannabis and cannabis derivatives as a treatment for pain.
                                        OUR REVIEW
We searched the English-language literature for studies published from 1960 to August 15,
2016 using electronic databases such as MEDLINE, EMBASE, CINAHL, EBM Reviews and
Cochrane Databases. Our search strategy includes the MESH terms: marijuana, cannabis,
cannabinoids, and pain. We also searched the bibliographies of systematic reviews and all
included studies by hand for additional relevant studies.
Eligibility criteria
We included all randomized controlled trials that compared cannabis to a control and examined
pain as an outcome; the control could be placebo or another analgesic. We excluded cross-
sectional studies, prospective and retrospective observational studies, case-control studies, case
reports, case series and review articles. We also excluded studies that were published in abstract
form only.
We reviewed titles and abstracts from the literature search to select articles that met our
eligibility criteria. Then, one of the authors abstracted data from the selected articles, including
demographic and clinical characteristics of the study, study duration, intervention duration, and
outcomes. During our review, we further excluded studies that were not blinded and studies
with sample size less than 10 when considering efficacy data. When considering long-term
safety data, we did include open label studies as there was not much long-term safety data.
                                           FINDINGS
A total of 4,472 articles were identified through the literature search. Of these, there were only
28 studies that satisfied our eligibility criteria (see Figure 1). An additional 5 studies (1-5) were
identified through hand search of bibliographies of review articles and included studies. After
excluding 5 studies that were not blinded (6-10) and 1 with small sample size (11), we included
a total of 27 articles (see Table 1).
     The majority of evidence on cannabis and pain comes from randomized controlled trials
that use either a placebo or known analgesics as controls. As there is no standard dose for
cannabinoids, the studies vary greatly in the amount of the cannabinoid administered. Specific
studies covered conditions that caused pain such as cancer, multiple sclerosis (MS), HIV-
induced neuropathy, fibromyalgia, or trauma-induced neuropathy. Other studies were more
                                     Cannabis and pain                                   157
general and did not specify the primary cause of pain. Of particular interest is the use of
cannabis for cancer pain and multiple sclerosis as cannabis is thought to have other positive
anti-emetic and anti-spastic effects. The primary methods of administering the treatments
included use of a nabiximol oromucosal spray, ingestion of cannabis extract capsules, and
inhalation of smoked cannabis.
                                      Mean
 First author,      No. of    Male               Clinical                      Duration
                                       age*                   Type of pain                   Treatment         Outcome          Results           Adverse events
 year, country     subjects   (%)               condition                      of study
                                       (SD)
Abrams et al.     55          87     48.5      HIV            Neuropathy      3 weeks     Smoked              Ratings of    Pain Reduction      -Anxiety, Sedation,
(2007), USA                          (6.6)                                                Cannabis (3.56%     pain (VAS)    by 34% using        Disorientation,
                                                                                          THC) x 3/ day                     Cannabis vs.        Paranoia,
                                                                                          for 5 days vs                     17% by placebo      Confusion,
                                                                                          placebo                           (p= 0.03)           Dizziness, and
                                                                                                                                                Nausea (all mild)
                                                                                                                                                -No dropouts due to
                                                                                                                                                adverse events
Berman et al.     48          96     39 (10)   Brachial       Neuropathy      6 weeks     Sativex Oro-        Pain on an    Pain Scores         Dizziness (n=9),
(2004), United                                 Plexus                                     mucosal Spray       11 point      -Baseline: 7.5, -   Somnolescence (n
Kingdom                                        Avulsion                                   (2.7 mg THC +       box scale,    After Placebo:      =7), Dysgeusia (n
                                                                                          2.5 mg CBD/         Short-Form    6.9, -After 1:1     =10), Nausea (n =
                                                                                          spray) vs placebo   McGill        THC:CBD: 6.1        1), Feeling Drunk
                                                                                          and THC Oro-        Pain          (p = 0.005),        (n = 4) (all mild to
                                                                                          mucosal Spray       Question-     -After THC          moderate)
                                                                                          (2.7 mg THC/        naire,        only: 6.3 (p =
                                                                                          spray) x 1-48       Pain          0.02)
                                                                                          sprays vs           Disability
                                                                                          placebo             Index
Cooper et al.     30          50     27 (6)    Cold Pressor   Nociceptive     5 x 6-7     Smoked              Pain on an    Unable to           Subjective Drug
(2013), USA                                    Test                           hour        Cannabis (1.98%     11 item       abstract due to     Effects: Smoked
                                                                              sessions    and 3.56%           numeric       method of           Cannabis (1.98%
                                                                                          THC),               scale         recording           and 3.56% THC)
                                                                                          Dronabinol 10                                         and 20 mg
                                                                                          mg, 20 mg,                                            Dronabinol
                                                                                          placebo                                               produced increased
                                                                                                                                                “High” and “Good”
                                                                                                                                                Drug Effects
                                                                                                                                                relative to placebo
Corey-Bloom et    30          37     51 (8)    Multiple       Spasticity      17 days     Smoked              Pain (using   Effect (95% CI)     Acute Cognitive
al. (2012), USA                                Sclerosis      (nociceptive)               Cannabis (4%        VAS)          = 5.28 (2.48 –      Effects (Dizziness,
                                                                                          THC) x 1/ day vs                  10.01)              Headache, Fatigue,
                                                                                          placebo                           reduction in        feeling too “high”)
                                                                                                                            VAS scores in
                                                                                                                            cannabis vs
                                                                                                                            placebo group
                                     Mean
 First author,     No. of    Male              Clinical                  Duration
                                      age*                Type of pain                Treatment        Outcome           Results          Adverse events
 year, country    subjects   (%)              condition                  of study
                                      (SD)
Ellis et al.     34          97     49.1     HIV          Neuropathy     7 weeks    Smoked            Pain           Median             -Concentration
(2012), USA                         (6.9)                                           Cannabis (1-8%    intensity on   difference in      Difficulties,
                                                                                    THC) x 4/day vs   a              DDS points for     Fatigue, Sleepiness,
                                                                                    placebo           Descriptor     pain reduction     Sedation, Increased
                                                                                                      Differen-      of cannabis vs.    sleep duration,
                                                                                                      tial Scale     placebo: 3.3 (p    Reduced salivation
                                                                                                      (DDS)          = 0.016)           and thirst; moderate
                                                                                                                                        to severe adverse
                                                                                                                                        events more
                                                                                                                                        common in
                                                                                                                                        treatment group -1
                                                                                                                                        drop out due to
                                                                                                                                        Psychosis, 1 due to
                                                                                                                                        Intractable Cough
Frank et al.     96          52     50.2     Various      Neuropathy     6 weeks    250ug-2 mg        Pain (using    VAS Scores         Tiredness (n = 79),
(2008), United                      (13.6)                                          Nabilone or 30-   VAS)           (mm):              Sleeplessness (n =
Kingdom                                                                             240 mg                           -Baseline: 69.6    46), Sickness
                                                                                    dihydrocodeine                   (range 29.4-       (n=46), Tingling (n
                                                                                    per day                          95.2), -           = 25), Strangeness
                                                                                                                     Nabilone: 59.      (n = 27),
                                                                                                                     93 (SD =           Nightmares (n = 7),
                                                                                                                     24.42), -          Shortness of Breath
                                                                                                                     Dihydrocodeine     (n=18), Headaches
                                                                                                                     : 58. 58 (SD =     (n = 20), Other (n =
                                                                                                                     24.08)             66)
                                                                                                                     -3/64 in
                                                                                                                     Nabilone group
                                                                                                                     had a clinically
                                                                                                                     significant drop
                                                                                                                     in VAS (by 10
                                                                                                                     mm or more)
Johnson et al.   177         54     60.2     Cancer       Mixed          2 weeks    Oromucosal        Pain on an     Numeric Pain       -THC:CBD:
(2010), United                      (12.3)                                          THC Extract       11 item        Scale Scores:      Somnolescence (n =
Kingdom                                                                             Spray (2.7 mg     numeric        -Baseline          8), Dizziness (n =
                                                                                    THC per spray),   scale          THC:CBD            7), Confusion
                                                                                    Oromucosal                       Group(SD):         (n=4), Nausea
                                                                                    THC:CBD                          5.68 (1. 24),      (n=6), Vomiting
                                                                                    Extract Spray,                   -Baseline THC      (n = 3), Raised
                                                       Table 1. (Continued)
                                  Mean
First author,    No. of    Male           Clinical                  Duration
                                  age*               Type of pain                 Treatment        Outcome      Results         Adverse events
year, country   subjects   (%)           condition                  of study
                                  (SD)
                                                                               (2.7 mg THC                   Group (SD):      Gamma GT (n =2),
                                                                               + 2.5 mg CBD                  5.77 (1.33),     Hypercalcemia (n =
                                                                               per spray) x 8-12             -Baseline        0), Hypotension (n
                                                                               sprays according              Placebo(SD):     =3) -THC:
                                                                               to individual                 6.05(1.32),      Somnolescence (n =
                                                                               response,                     -Median          8), Dizziness (n =
                                                                               placebo                       Reduction        7), Confusion
                                                                                                             THC:CBD          (n=1), Nausea (n =
                                                                                                             Group: -         4), Vomiting (n =
                                                                                                             1.37(p=0.024),   4), Raised Gamma
                                                                                                             -Median          GT (n = 5), -Mostly
                                                                                                             Reduction THC    mild or moderate.
                                                                                                             Group: -
                                                                                                             1.01(p=0.204),
                                                                                                             -Median
                                                                                                             Reduction
                                                                                                             Placebo Group:
                                                                                                             -0.69
                                                                                                             -Statistically
                                                                                                             significant
                                                                                                             reduction for
                                                                                                             THC:CBD
                                                                                                             group only,
                                                                                                             clinically
                                                                                                             significant
                                                                                                             (>30% pain
                                                                                                             Reduction)
                                                                                                             reduction
                                                                                                             observed in
                                                                                                             43% of
                                                                                                             treatment
                                                                                                             patients vs.
                                                                                                             21% of placebo
                                                                                                             patients
                                        Mean
 First author,        No. of    Male                Clinical                       Duration
                                         age*                  Type of pain                        Treatment         Outcome          Results          Adverse events
 year, country       subjects   (%)                condition                       of study
                                         (SD)
Kalliomaki et al.   30          100    29.3       Healthy      Nociceptive        3 treatment   Nabilone            Pain (using   No significant     -Dizziness (n = 21),
(2013), Sweden                         (6.25)     volunteers   using a heat-      sessions      Capsules (1 mg,     a VAS)        difference         Postural Dizziness
                                                               pain model or                    2 mg, and 3 mg),                  between            (n = 11), Fatigue (n
                                                               intradermal                      placebo. Single                   nabilone doses     = 10), Dry Mouth
                                                               capsaicin                        dose.                             and placebo for    (n=10),
                                                                                                                                  electronic VAS     Tachycardia (n =
                                                                                                                                  (maximal pain)     9). Mostly mild to
                                                                                                                                  or VAS (area       moderate.
                                                                                                                                  under curve)       - 14 severe adverse
                                                                                                                                                     events led to the
                                                                                                                                                     withdrawal of four
                                                                                                                                                     patients
Killestein et al.   16          N/A    46 (7.9)   Multiple     Spasticity         12 weeks      Dronabinol          Pain (using   No abstractable    -Adverse events
(2002), Nether-                                   Sclerosis    (nociceptive)                    (THC) 2.5 mg,       VAS)          pain data          were more common
lands                                                                                           C. sativa extract                                    with the C. sativa
                                                                                                (2.5 mg THC,                                         extract than the
                                                                                                CBD, other                                           Dronabinol
                                                                                                cannabinoids)                                        treatment. All were
                                                                                                                                                     mild to moderate.
                                                                                                                                                     -1 severe adverse
                                                                                                                                                     effect (acute
                                                                                                                                                     psychosis) with C.
                                                                                                                                                     sativa extract.
Kraft et al.        18          0      23.5       Healthy      Nocioceptive--     8 hour        Oral Cannabis       Pain (using   No significant     Drowsiness,
(2008),                                (2.6)      volunteers   Sunburn            sessions      Extract Capsules    VAS)          difference         Sedation, Dry
Germany                                                        Hyperalgesia,                    (2:1 THC:CBD                      between            Mouth, and Vertigo
                                                               Electrical Pain,                 Content) (20 mg                   cannabis extract   had significantly
                                                               and                              THC) per session                  capsules and       different self-report
                                                               Intradermal                                                        placebo was        and observer
                                                               Capsaicin                                                          found for any      reported VAS
                                                                                                                                  pain models        scores for the
                                                                                                                                                     cannabis treatment
                                                                                                                                                     compared to
                                                                                                                                                     placebo
                                                               Table 1. (Continued)
                                        Mean
 First author,       No. of    Male               Clinical                   Duration
                                         age*                Type of pain                   Treatment         Outcome           Results         Adverse events
 year, country      subjects   (%)               condition                   of study
                                         (SD)
Langford           339         32      49.0     Multiple     Neuropathy     (Phase A) 4   Oral muscosa       Validated      Responders at     15 (9%) in the
(2013), 33 study                       (10.5)   sclerosis                   weeks         spray (2.7 mg of   self-          the 30 %          THC/CBD spray
sites globally                                                                            THC + 2.5 mg of    reported 0–    improvement       group and 12 (7 %)
                                                                                          CBD), x max 12     10 point       level in mean     in the placebo
                                                                                          sprays/ day,       numerical      pain NRS score    group, stopped
                                                                                          placebo            rating scale   totaled 50 % in   study medication
                                                                                                             (NRS)          the THC/CBD       due to AEs.
                                                                                                             assessing      spray group vs    Majority of AEs
                                                                                                             mean daily     45 % in           leading to
                                                                                                             neuro-         the placebo       permanent cessation
                                                                                                             pathy          group             of study medication
                                                                                                                            (p = 0.234)       were within the
                                                                                                                                              nervous system
                                                                                                                                              disorders and
                                                                                                                                              gastrointestinal
                                                                                                                                              disorders system
                                                                                                                                              organ classes
Lynch et al.       18          17/83   56       Chemothera   Neuropathy     6 months      Oromucosal         Pain on an     No significant    Fatigue (n = 7), Dry
(2014), United                         (10.8)   py induced                                THC Extract        11 item        difference was    Mouth (n = 5),
Kingdom                                         pain                                      Spray (2.7 mg      numeric        found between     Dizziness (n = 6),
                                                                                          THC/spray),        scale          placebo and the   Nausea (n = 6),
                                                                                          Oromucosal                        nabiximol         Increased Appetite
                                                                                          THC:CBD                           spray, however    (n = 2), Diarrhea (n
                                                                                          Extract Spray,                    5/16              = 2), Decreased
                                                                                          (2.7 mg THC +                     participants      Appetite (n = 1),
                                                                                          2.5 mg CBD/                       experienced a     Feeling “stoned” (n
                                                                                          spray) x 1-12                     clinically        = 1), Anxiety (n =
                                                                                          sprays according                  significant       1), Panic Attack (n
                                                                                          to individual                     reduction in      = 1), Headache (n =
                                                                                          response,                         pain at the end   2), Confusion (n =
                                                                                          placebo                           of 6 months.      1), “Fuzzy
                                                                                                                                              Thinking” or
                                                                                                                                              “Foggy Brain” (n =
                                                                                                                                              1)
                                       Mean
 First author,      No. of    Male                Clinical                     Duration
                                       age*                    Type of pain                  Treatment         Outcome          Results          Adverse events
 year, country     subjects   (%)                condition                     of study
                                       (SD)
Narang (2008),    30          47     Median     Non-cancer     Chronic pain   Phase 1 ~4   10 mg or 20 mg     Total Pain    -Patients on       2 adverse events
USA                                  (range):   pain (taking                  weeks        of dronabinol      Relief at 8   Dronabinol had     during the study,
                                     43.5       opiods)                                    (single dose),     hours         decreased pain     both related to
                                     (21-67)                                               placebo            (TOTPAR)      intensity and      anxiety (tremors,
                                                                                                                            increased          dizziness, inability
                                                                                                                            satisfaction       to concentrate).
                                                                                                                            compared with      Both occurred in
                                                                                                                            placebo.           subjects who
                                                                                                                            -No difference     received
                                                                                                                            in benefit were    20 mg of
                                                                                                                            found between      dronabinol.
                                                                                                                            the 20 mg and
                                                                                                                            10 mg
Nurmikko et al.   125         41     53.3       Allodynia      Neuropathy     5 weeks      Sativex            Pain on an    -Difference        -Dizziness (n = 18),
(2007), United                       (15.5)                                                Oromucosal         11 item       between            Nausea (n = 14),
Kingdom                                                                                    Spray (2.7 mg      numeric       Sativex and        Fatigue (n = 13),
                                                                                           THC + 2.5 mg       scale,        Placebo            Dry Mouth (n =
                                                                                           CBD/ spray) x 1-   Neuropathi    Decrease: -0.96    11), Vomiting (n =
                                                                                           48 sprays          c Pain        (p=0.004)          8), Feeling Drunk
                                                                                           according to       Scale         -Statistically     (n = 6), Headache
                                                                                           individual                       significant        (n = 6), Diarrhea (n
                                                                                           response,                        decrease in pain   = 4),
                                                                                           placebo                          -26% of            Nasopharyngitis (n
                                                                                                                            Sativex vs 15%     = 4), Anorexia (n =
                                                                                                                            placebo patients   4), Somnolescence
                                                                                                                            achieved a         (n = 4), Abdominal
                                                                                                                            clinically         Pain Upper (n=3),
                                                                                                                            significant        Disturbance in
                                                                                                                            decrease in pain   Attention (n=3),
                                                                                                                            (>30% pain         Memory
                                                                                                                            reduction)         Impairmnet (n = 3)
                                                                                                                                               -91% of patients
                                                                                                                                               experienced at
                                                                                                                                               least1 AE; most
                                                                                                                                               were mild
                                                             Table 1. (Continued)
                                      Mean
 First author,      No. of    Male              Clinical                  Duration
                                      age*                 Type of pain                 Treatment         Outcome         Results           Adverse events
 year, country     subjects   (%)              condition                  of study
                                      (SD)
                                                                                                                                          -11% on Sativex
                                                                                                                                          experienced a
                                                                                                                                          severe AE vs. 8%
                                                                                                                                          on placebo
Portenoy et al.   360         52     58       Various      Various        24 months   Sativex            Pain on an   No statistical or   Neoplasm
(2012), US                           (12.2)                                           Oromucosal         11 item      clinical            Progression (n =
                                                                                      Spray (2.7 mg      numeric      significant         47), Nausea (n =
                                                                                      THC + 2.5 mg       scale        difference in       59), Dizziness (n
                                                                                      CBD/ spray) x 1-                pain relief         =51), Vomiting (n
                                                                                      4,6-10,11-16                    between             =42),
                                                                                      sprays according                Sativex spray       Somnolescence
                                                                                      to individual                   and placebo         (n=39),
                                                                                      response                                            Disorientation (n =
                                                                                                                                          18), Anorexia (n =
                                                                                                                                          22), Constipation
                                                                                                                                          (n=20), Dry Mouth
                                                                                                                                          (n=22), Anemia
                                                                                                                                          (n=19), Diarrhea
                                                                                                                                          (n=17), Dysgeusia
                                                                                                                                          (n = 11), Headache
                                                                                                                                          (n =15), Asthenia (n
                                                                                                                                          =18),
                                                                                                                                          Hallucination (n =
                                                                                                                                          8), Decreased
                                                                                                                                          Appetite (n = 11),
                                                                                                                                          Fatigue (n = 13),
                                                                                                                                          Pain (n = 11),
                                                                                                                                          Insomnia (n = 8),
                                                                                                                                          Stomatitis (n =11),
                                                                                                                                          Weight Decreased
                                                                                                                                          (n = 8)
                                                                                                                                          -29.5% of patients
                                                                                                                                          receiving Sativex
                                                                                                                                          had a severe
                                                                                                                                          adverse event
                                     Mean
 First author,     No. of    Male              Clinical                   Duration
                                      age*                 Type of pain                Treatment         Outcome          Results          Adverse events
 year, country    subjects   (%)              condition                   of study
                                      (SD)
Rog et al.       66          21     49.2     Multiple      central pain   4 weeks    Sativex            Pain on an    Numeric Pain       -Dizziness (n = 18),
(2005), United                      (8.3)    Sclerosis                               Oromucosal         11 item       Scale Scores       Somnolescence (n =
Kingdom                                                                              Spray (2.7 mg      numeric       (SD) -Baseline     3), Disturbance in
                                                                                     THC + 2.5 mg       scale         Sativex: 6.5       Attention (n = 2),
                                                                                     CBD/ spray) x 1-                 (1.6), -Baseline   Headache (n = 1),
                                                                                     48 sprays                        Placebo: 6.4       Dissociation (n =
                                                                                     according to                     (1.7),-Decrease    3), Euphoria (n =
                                                                                     individual                       in Pain Scale      2), Dry Mouth (n =
                                                                                     response,                        Score Sativex,     4), Nausea (n = 3),
                                                                                     placebo                          Placebo: -2.7, -   Diarrhea (n = 2),
                                                                                                                      1.4 (between       Glossodynia (n =
                                                                                                                      groups             1), Mouth
                                                                                                                      difference((p =    Ulceration
                                                                                                                      0.005)             (n = 1), Vomiting (n
                                                                                                                                         = 1), Falls (n = 3),
                                                                                                                                         Weakness (n = 3),
                                                                                                                                         Fatigue (n = 2),
                                                                                                                                         Feeling
                                                                                                                                         Abnormal (n = 1),
                                                                                                                                         Feeling Drunk (n
                                                                                                                                         =1), Pharyngitis (n
                                                                                                                                         = 2), Hoarseness (n
                                                                                                                                         = 1), Throat
                                                                                                                                         Irritation (n = 1)
                                                                                                                                         - 88.2% of patients
                                                                                                                                         on Sativex
                                                                                                                                         developed one or
                                                                                                                                         more AE,
                                                                                                                                         -2 patients
                                                                                                                                         withdrew due to
                                                                                                                                         severe AEs
Skrabek et al.   40          7      48.9     Fibromyalgi   Nociceptive    8 weeks    Nabilone (THC)     Pain (using   -Decrease in       After 4 weeks:
(2008), Canada                      (7.6)    a                                       Capsules (0.5-1    VAS)          Pain for 1 mg      -Drowsiness (n =
                                                                                     mg)/day for one                  Nabilone BID       7), Dry Mouth (n =
                                                                                     week?, placebo                   from Baseline      5), Vertigo (n = 4),
                                                                                                                      after 4 weeks:     Ataxia (n = 3),
                                                                                                                       -2.04 (p <        Confusion (n = 2),
                                                                                                                      0.02).             Decreased
                                                              Table 1. (Continued)
                                      Mean
 First author,      No. of    Male              Clinical                   Duration
                                      age*                 Type of pain                 Treatment       Outcome         Results          Adverse events
 year, country     subjects   (%)              condition                   of study
                                      (SD)
                                                                                                                    -No significant    Concentration (n =
                                                                                                                    differences        2), Dissociation (n
                                                                                                                    observed for       = 2), Orthostatic
                                                                                                                    0.5 mg             Hypertension (n=2),
                                                                                                                    Nabilone PO at     Anorexia (n = 2),
                                                                                                                    2 or 4 weeks       Headache (n = 1),
                                                                                                                    and no             Dysphoria (n = 1),
                                                                                                                    significant        Euphoria (n = 1),
                                                                                                                    differences        Sensory
                                                                                                                    observed for 1     Disturbance (n = 1).
                                                                                                                    mg Nabilone        Mostly mild.
                                                                                                                    BID at 2 weeks.    -No serious adverse
                                                                                                                                       events occurred
                                                                                                                                       -Adverse events
                                                                                                                                       were significantly
                                                                                                                                       more common in
                                                                                                                                       the treatment group
                                                                                                                                       at 2 and 4 weeks
Svendson et al.   24          42     50 (8)   Multiple     Spasticity      6 weeks    Dronabinol       Pain on an   -Median            -Dizziness or
(2004),                                       Sclerosis    (nociceptive)              (THC) 10         11 item      spontaneous        Lightheadedness (n
Denmark                                                                               mg/day for 3     numeric      pain               = 14), Tiredness or
                                                                                      weeks, placebo   scale        significantly      Drowsiness (n =
                                                                                                                    lower for          10), Fatigue (n = 1),
                                                                                                                    Dronabinol         Balance Difficulty
                                                                                                                    than for           (n = 2), Headache
                                                                                                                    placebo            (n = 6), Migraine (n
                                                                                                                    (p=0.02) with      = 1), Speech
                                                                                                                    13 (54%)           Disorders (n = 1),
                                                                                                                    patients           Feeling of
                                                                                                                    achieving          Drunkenness (n =
                                                                                                                    clinically         2), Sleep Difficulty
                                                                                                                    significant        (n = 1), Multiple
                                                                                                                    (>30% pain         Sclerosis
                                                                                                                    reduction) pain    Aggravated (n = 1),
                                                                                                                    reduction on       Myalgia (n = 6),
                                                                                                                    active treatment   Muscle Weakness
                                     Mean
 First author,     No. of    Male              Clinical                   Duration
                                     age*                  Type of pain                Treatment         Outcome           Results          Adverse events
 year, country    subjects   (%)              condition                   of study
                                     (SD)
                                                                                                                       versus 5 (21%)     (n = 3), Limb
                                                                                                                       on placebo         Heaviness (n = 1),
                                                                                                                                          Distortion of Wrist
                                                                                                                                          (n = 1), Mouth
                                                                                                                                          Dryness (n = 3),
                                                                                                                                          Nausea (n = 3),
                                                                                                                                          Palpitations (n = 4),
                                                                                                                                          Euphoria (n = 3),
                                                                                                                                          Hyperactivity (n =
                                                                                                                                          1), Hot Flushes (n =
                                                                                                                                          1), Anorexia (n =
                                                                                                                                          1), Chills (n = 1),
                                                                                                                                          Upper Airway
                                                                                                                                          Infection (n = 1),
                                                                                                                                          Tenderness in Nose
                                                                                                                                          (n = 1)
                                                                                                                                          -Number of adverse
                                                                                                                                          events was high for
                                                                                                                                          the active treatment
                                                                                                                                          group than for the
                                                                                                                                          placebo group
                                                                                                                                          -No severe adverse
                                                                                                                                          events were
                                                                                                                                          reported; no
                                                                                                                                          withdrawals due to
                                                                                                                                          adverse events
Toth (2012),     26          54     61.2     Refractory    Neuropathy     5 weeks    nabilone 1-4       Pain diaries   Improvement in     Treatment-emergent
Canada                              (14.7)   human                                   mg/day, taken as                  the change in      adverse
                                             diabetic                                2/day, 12 hours                   mean end-point     events were
                                             peripheral                              apart                             neuropathic        reported by 6/13
                                             neuropathic                                                               pain in nabilone   (46%) subjects
                                             pain                                                                      vs                 receiving placebo
                                                                                                                       placebo (mean      and by 7/13 (54%)
                                                                                                                       treatment          subjects receiving
                                                                                                                       reduction of       nabilone (most
                                                                                                                       1.27; 95%          were mild or
                                                                                                                       confidence         moderate)
                                                                                                                       interval 2.29-
                                                                                                                       0.25)
                                                                Table 1. (Continued)
                                       Mean
 First author,       No. of    Male              Clinical                    Duration
                                        age*                  Type of pain                 Treatment       Outcome          Results          Adverse events
 year, country      subjects   (%)              condition                    of study
                                        (SD)
Turcotte (2015),   15          13     45.5     Relapsingre    MS-induced     9 weeks    Nabilone          Pain (using   After              Nabilone
Canada                                (10.8)   mitting        neuropathic               5-week            VAS): pain    adjustment, a      was well tolerated,
                                               multiple       pain                      maintenance of    intensity     significant        with dizziness/
                                               sclerosis                                1mg oral          (VASpain),    group x time2      drowsiness
                                               (MS)                                     nabilone          and           interaction term   most frequently
                                               patients (on                             (placebo) twice   impact of     was reported       reported.
                                               gabapentin)                              daily             pain on       for both the
                                                                                                          daily         VASpain and
                                                                                                          activities    VASimpact
                                                                                                          (VAS          score,
                                                                                                          impact)       demonstrating
                                                                                                                        the adjusted
                                                                                                                        rate of decrease
                                                                                                                        for both
                                                                                                                        outcomes was
                                                                                                                        statistically
                                                                                                                        greater in
                                                                                                                        nabilone vs
                                                                                                                        placebo group.
Wade et al.        24          50%    N/A      Various        Various        8 weeks    Sativex           Pain (using   - Statistically    -THC:CBD only:
(2003), United                                                                          Oromucosal        VAS)          significant        “Drug Toxicity” (n
Kingdom                                                                                 Spray (2.7 mg                   decrease in pain   = 1), Headache (n =
                                                                                        THC + 2.5 mg                    for THC and        1), Nausea (n = 1),
                                                                                        CBD/ spray),                    CBD                Vomiting (n = 1),
                                                                                        THC                             treatments, but    Diarrhea (n = 1),
                                                                                        Oromucosal                      not THC:CBD        Sleepiness (n = 2),
                                                                                        Spray (2.7 mg                   treatment          Fall (n = 1), Cough
                                                                                        THC), CBD                                          (n = 1), Impaired
                                                                                        Oromucosal                                         Balance (n = 1),
                                                                                        Spray (2.5 mg                                      Fatigue (n = 1),
                                                                                        CBD) x 1-44                                        Influenza-like
                                                                                        (120 mg                                            Symptoms (n = 1),
                                                                                        THC/120 mg                                         Thirst (n = 1)
                                                                                        CBD) sprays                                        -Feeling Intoxicated
                                                                                        according to                                       led to 3 withdrawals
                                                                                        individual
                                     Mean
 First author,     No. of    Male              Clinical                    Duration
                                     age*                 Type of pain                  Treatment      Outcome          Results          Adverse events
 year, country    subjects   (%)              condition                    of study
                                     (SD)
                                                                                      response,
                                                                                      placebo
Wallace et al.   19          58     28.9     Healthy      Nociceptive –   1 day       Smoked          Pain (using   Five minutes       -Low Dose:
(2007), USA                         (10.9)   volunteers   pain induced                Cannabis (2%,   VAS;          after cannabis     Dizziness/Faintness
                                                          by Capsaicin                4%, 8% THC)     Visual        exposure, there    (n = 1), Injection
                                                                                                      Analogue      was no             Side Effects (n = 2).
                                                                                                      Spon-         effect on          -No Medium Dose
                                                                                                      taneous       capsaicin-         Adverse
                                                                                                      Pain          induced pain at    Events reported.
                                                                                                      Intensity -   any dose. By 45    - High Dose:
                                                                                                      VASPI)        min after          Dizziness/Faintness
                                                                                                                    cannabis           (n = 3),
                                                                                                                    exposure,          Somnolescence (n =
                                                                                                                    however, there     1), Feeling Cold (n
                                                                                                                    was a              = 1), Cognitive
                                                                                                                    significant        Impairment (n = 1),
                                                                                                                    decrease           Dyspnea (n = 1),
                                                                                                                    in capsaicin-      Dry Mouth (n = 1),
                                                                                                                    induced pain       Injection Side
                                                                                                                    with the           Effects (n = 1),
                                                                                                                    medium dose        Nausea and
                                                                                                                    and a              Vomiting (n = 1)
                                                                                                                    significant
                                                                                                                    increase in
                                                                                                                    capsaicin-
                                                                                                                    induced pain
                                                                                                                    with the high
                                                                                                                    dose.
                                                                                                                    There was no
                                                                                                                    effect seen with
                                                                                                                    the low dose,
                                                                                                                    nor was there
                                                                                                                    an effect on the
                                                                                                                    area of
                                                                                                                    hyperalgesia at
                                                                                                                    any dose.
                                                                                                                    Significant
                                                                                                                    negative
                                                                                                                    correlations
                                                             Table 1. (Continued)
                                     Mean
 First author,     No. of    Male              Clinical                   Duration
                                     age*                  Type of pain                Treatment       Outcome         Results          Adverse events
 year, country    subjects   (%)              condition                   of study
                                     (SD)
                                                                                                                   between pain
                                                                                                                   perception and
                                                                                                                   plasma _-9-
                                                                                                                   tetrahydrocanna
                                                                                                                   binol levels
                                                                                                                   were found
                                                                                                                   after adjusting
                                                                                                                   for the
                                                                                                                   overall dose
                                                                                                                   effects.
Ware et al.      23          48     45.4     Post-         Neuropathy     8 weeks    Smoked           Pain on an   Difference         -9.4% THC:
(2010), Canada                      (12.3)   Trauma/Surg                             Cannabis         11 item      between            Asthenia (n = 2),
                                             ery                                     (2.5%,6%, and    numeric      placebo and        Decreased Motor
                                                                                     9.4% THC) x 3/   scale        9.4% THC: 0.7      Skill (n = 1),
                                                                                     day, placebo                  (p=0.023)          Dizziness (n = 4),
                                                                                                                   -Statistically     Headache (n = 4),
                                                                                                                   significant pain   Heavy-headed (n =
                                                                                                                   relief for 9.4%    1), Lightheaded (n
                                                                                                                   THC cigarettes,    = 1), Numbness (n
                                                                                                                   but non-           = 2), Sleepiness (n
                                                                                                                   significant pain   = 2), Unbalanced (n
                                                                                                                   relief at other    = 1), Burning
                                                                                                                   THC                Sensation (n = 3),
                                                                                                                   concentrations     Fatigue (n = 1),
                                                                                                                                      Heaviness (n = 1),
                                                                                                                                      Hematoma (n = 1),
                                                                                                                                      Irritation of Oral
                                                                                                                                      Cavities (n = 1),
                                                                                                                                      Itchiness (n = 1),
                                                                                                                                      Itchiness in Face (n
                                                                                                                                      = 1), Itchiness of
                                                                                                                                      Nose (n = 1), Pain
                                                                                                                                      (n = 2), Tingling
                                                                                                                                      Nose (n = 1),
                                                                                                                                      Craving for Sweets
                                                                                                                                      (n = 1), Disinterest
                                     Mean
 First author,     No. of    Male    age*        Clinical                    Duration
                                                             Type of pain                  Treatment       Outcome          Results       Adverse events
 year, country    subjects   (%)     (SD)       condition                    of study
                                                                                                                                        in Surroundings (n
                                                                                                                                        = 1), Dysphoria (n
                                                                                                                                        = 2), Euphoria (n =
                                                                                                                                        1), Fidgety Fingers
                                                                                                                                        (n = 1), Foggy
                                                                                                                                        Mental State (n =
                                                                                                                                        1), Lack of
                                                                                                                                        Concentration (n =
                                                                                                                                        2), Less Alert (n =
                                                                                                                                        1), Paranoia (n = 1),
                                                                                                                                        Racing Thoughts (n
                                                                                                                                        = 1), Cough (n = 3),
                                                                                                                                        Short of Breathe (n
                                                                                                                                        = 1), Throat
                                                                                                                                        Irritation (n = 3),
                                                                                                                                        Dry Mouth (n = 1),
                                                                                                                                        Increased Appetite
                                                                                                                                        (n = 2), Nausea (n =
                                                                                                                                        1), Dry Eyes (n =
                                                                                                                                        1), Itchiness of Eyes
                                                                                                                                        (n = 1), Edema (n =
                                                                                                                                        1), Injury to Right
                                                                                                                                        Knee (n = 1); all
                                                                                                                                        were mild to
                                                                                                                                        moderate.
                                                                                                                                        -No severe adverse
                                                                                                                                        events
Wilsey et al.    38          53     Median     Unspecified   Neuropathy     3 x 6 hour   Smoked           Pain (using   -Mean           Adverse events
(2008), USA                         (range):                                sessions     Cannabis (3.5%   VAS) and      Difference      were not mentioned
                                    46 (21–                                              and 7% THC) x    Neuro-        VAS Pain        individually,
                                    71)                                                  9 puffs per      pathic Pain   Intensity per   however a
                                                                                         session          Scale         minute (7%      significant “good
                                                                                                                        THC vs.         drug effect” was
                                                                                                                        Placebo): -     reported for both
                                                                                                                        0.0035 (p =     treatments when
                                                                                                                        0.04),          compared to the
                                                                                                                        -Mean           placebo and a
                                                              Table 1. (Continued)
                                     Mean
 First author,     No. of    Male               Clinical                    Duration
                                     age*                   Type of pain                  Treatment         Outcome          Results          Adverse events
 year, country    subjects   (%)               condition                    of study
                                     (SD)
                                                                                                                         Difference         significant “bad
                                                                                                                         VAS Pain           drug effect” was
                                                                                                                         Intensity per      reported only for
                                                                                                                         minute (3.5%       the 7% THC dose
                                                                                                                         THC vs.            nearer to the end of
                                                                                                                         Placebo):          the trial
                                                                                                                          -0.0036
                                                                                                                         (p=0.03)
                                                                                                                         -No significant
                                                                                                                         difference
                                                                                                                         between the
                                                                                                                         two THC doses
Wilsey et al.    39          72     50 (11)   Unspecified   Neuropathy     3 x 6 hour   Smoked             Pain (using   -Difference        Adverse events
(2013), USA                                                                sessions     Cannabis (1.29%    VAS) and      between both       were not mentioned
                                                                                        and 3.5% THC)      Neuropathi    treatment          individually,
                                                                                        x 8-12 puffs per   c Pain        groups and         however a
                                                                                        session            Scale         placebo            significant “good
                                                                                                                         observed at 120    drug effect” was
                                                                                                                         mins (p =          noted with both
                                                                                                                         0.0002), and       treatments when
                                                                                                                         evident at 240     compared to
                                                                                                                         mins (p =          placebo and a
                                                                                                                         0.0004), and       significant “bad
                                                                                                                         300 mins (p =      drug effect” was
                                                                                                                         0.018).            noted with the
                                                                                                                         -10/38 patients    medium dose THC
                                                                                                                         had a clinically   at 240 mins
                                                                                                                         significant
                                                                                                                         (>30% pain
                                                                                                                         reduction)
                                                                                                                         reduction in
                                                                                                                         pain on placebo
                                                                                                                         vs. 21/37 on the
                                                                                                                         low dose THC
                                                                                                                         and 22/ 36 on
                                       Mean
  First author,      No. of    Male              Clinical                   Duration
                                       age*                 Type of pain                  Treatment      Outcome           Results          Adverse events
  year, country     subjects   (%)              condition                   of study
                                       (SD)
                                                                                                                       the medium
                                                                                                                       dose THC.
 Wissel et al.     17          24     44.8     Multiple     Spasticity      9 weeks    Nabilone         Pain on an     Nabilone           1 patient dropped
 (2006), Austria                      (14.4)   Sclerosis    (Nociceptive)              Capsules (1 mg   11 item        treatment          out due to weakness
                                                                                       per day)         numeric        decreased by a     of the lower limbs,
                                                                                                        scale,         median 2 points    other adverse events
                                                                                                        Ashworth       when compared      not mentioned
                                                                                                        Score          to placebo (p <
                                                                                                        (Spasticity)   0.05)
 Zajicek et al.    277         37     51.9     Multiple     Spasticity      12 weeks   Dronabinol       Pain on an     -Mean Change       -Dizziness (n = 89),
 (2012), United                       (7.8)    Sclerosis    (nociceptive)              (THC) 2.5 mg     11 item        in Body Pain       Urinary Tract
 Kingdom                                                                               taken up to 25   numeric        from Baseline      Infection(n = 34),
                                                                                       mg daily         Likert         Dronabinol         Dry Mouth (n =
                                                                                                        scale,         (SD): -1.2 (2.6)   34), Headache (n =
                                                                                                        Ashworth       -Mean Change       22), Asthenia (n =
                                                                                                        Score          in Body Pain       25), Fatigue (n =
                                                                                                        (Spasticity)   from Baseline      25).
                                                                                                        Pain is a      Placebo(SD):       -Mostly mild to
                                                                                                        secondary       -0.3 (2.4)        moderate.
                                                                                                        outcome.       -Statistically     -233/277
                                                                                                                       significant        experienced at least
                                                                                                                       decrease in pain   one adverse event
                                                                                                                       between THC        -More adverse
                                                                                                                       and placebo (p     events present in
                                                                                                                       <0.025)            the treatment group
                                                                                                                                          (93.0% vs. 74.6%)-
                                                                                                                                          30 patients
                                                                                                                                          withdrew from
                                                                                                                                          active treatment due
                                                                                                                                          to AE.
                                                                                                                                          -7 serious adverse
                                                                                                                                          events
VAS = Visual Analog Scale.
* When means and SDs are given for men and women separately, we combined the means and SDs using methods described in
   http://handbook.cochrane.org/chapter_7/table_7_7_a_formulae_for_combining_groups.htm.
174                             Jonathan K Hwang and Hance Clarke
Nabiximols are a cannabis extract that contain tetrahydrocannabinol (THC) and cannabinidiol
(CBD) in an approximate 1:1 ratio and are administered as a sublingual spray. This mix of
cannabinoids is marketed under the name Sativex in Europe and Canada, and contains 2.7 mg
THC and 2.5 mg CBD per spray. Studies that administered this spray almost exclusively left
dosing up to the patient, providing them with a range of allowed sprays per day, but not giving
them an exact dose that they should take in a period of time (2, 3, 5, 6, 12-16). As such, the
amount of THC and CBD consumed between individuals varied greatly.
     Nabiximols have been studied for their efficacy on pain as a result of chemotherapy-
induced neuropathy (13), cancer (12), multiple sclerosis (5, 15, 16), neuropathy characterized
by allodynia (2), various cancers (14), and neuropathy as a result of a brachial plexus avulsion
(3). Of the eight studies, five of the eight reported a statistically significant decrease in pain (2,
3, 12, 14, 15). Of the five studies that reported a decrease in pain, one showed quantitative
clinical relevance (12), another two did not show clinical relevance (3, 14) and two did not
mention clinical relevance in a quantitative form, if at all (2, 15). Side effects such as dry mouth,
dizziness, nausea, vomiting and fatigue were generally well tolerated, with relatively few severe
adverse events (3, 5, 12, 13, 15); however in two other studies, severe adverse events were
relatively common (29.5% of patients in one study (14) and 10% of patients in another study
(16)).
     The literature is sparse in terms of the long term effects of using nabiximol sprays. Two
open label studies, one documenting nabiximol use over 38 weeks for neuropathic pain (6) and
the other documenting nabiximol use over 2 years for terminal cancer pain (7), both concluded
that there were no significant adverse effects for long term usage, however more research needs
to be done to verify this finding. One study found that nabiximols were associated with a larger
incidence of severe adverse affects (29.5% of nabiximol treated patients vs. 24.2% of placebo
treated patients) and a larger incidence of death (20.9% of nabiximol treated patients vs. 17.6%
of placebo treated patients) over a period of 24 months (14). The study acknowledged that the
lack of correlation between severe adverse events, death, and number of Nabiximol sprays
administered means that the association likely came about purely by chance (14). Nevertheless,
more research is needed to examine this association.
     Overall, nabiximol sprays seem to have a statistically significant effect on pain, with
variable clinical relevance. A consideration with respect to studies of nabiximols is that the
majority of these studies were funded by the pharmaceutical company that manufactures this
product (2, 3, 5-7, 12, 15, 16).
Orally ingested capsules of cannabinoid extracts dronabinol and nabilone have been tested for
their analgesic effects. Dronabinol is a THC extract, while nabilone is a synthetic cannabinoid
that mimics the action of THC. Studies testing dronabinol varied in their dosing considerably,
giving dosages ranging from 2.5 mg to 25 mg per day (1, 4, 17-19), while studies testing
nabilone prescribed between 0.5 mg to 4 mg per day (20-25).
     Dronabinol was studied for its efficacy on pain as the result of multiple sclerosis (1, 4, 18),
unspecified chronic pain (19), and sunburn induced hyperalgesia (17). Three of the five studies
                                     Cannabis and pain                                         175
reported a statistically significant decrease in pain (4, 18, 19). Of these three studies, one study
claimed a 54% clinically significant decrease in pain with the active agent compared to a 21%
clinically significant decrease in pain with the placebo (4), while the other two studies did not
talk about clinical significance (18, 19). Of the two studies that did not find a decrease in pain,
one argued that dronabinol had no analgesic effects on acute inflammatory pain whatsoever
(17), while the other stated that although dronabinol use was found to be tolerable in patients,
its efficacy was questionable (1).
     Nabilone was studied for its efficacy on pain as a result of fibromyalgia (24), multiple
sclerosis (22, 25), chronic neuropathic pain (20), diabetic peripheral neuropathic pain (21), and
capsaicin induced pain (23). Five of the six studies for nabilone and pain reported a statistically
significant decrease in pain (20-22, 24, 25). Of the five studies, two discussed clinical
significance in quantitative terms. One of the studies reported that 3 of 64 patients experienced
a clinically significant decrease in pain (20) and the other study reported that 11 of 13 patients
experienced a clinically significant decrease in pain with nabilone compared to 5 of 13 on
placebo (21). All of the five studies concluded that nabilone was a tolerable and possibly
effective treatment (20-22, 24, 25). When nabilone was compared to a dihydrocodeine, it was
found to have less of an analgesic effect (20). The one study that did not find a significant
decrease in pain tested for the effects of a single dose of nabilone and acknowledged that this
finding could not rule out efficacy over a longer treatment period (23).
     In terms of adverse effects, studies identified certain short-term adverse effects (1, 4, 17-
25). The most common adverse events were tiredness, dizziness, lightheadedness, and
headaches, (1, 4, 17, 18, 20, 23-25) and some participants withdrew from studies due to these
events (18, 23, 24, 26). A study on the psychoactive effects of dronabinol found that its adverse
effects were similar to those of smoking cannabis (26). There were no studies that documented
the effects of using nabilone or dronabinol on a long-term basis.
     Overall, nabilone and dronabinol seem to have some analgesic effect on chronic pain. As
the extent to which the analgesic effect is clinically significant is highly debatable, more
research needs to be done to verify their effects. Despite their effect on chronic pain, cannabis
extract capsules may not have much of an effect on acute inflammatory pain. The current
literature on orally consumed cannabis extract capsules is sparse and more research is required
to draw conclusions on their effect on pain.
Smoked cannabis
Smoking cannabis is the simplest way of ingesting cannabis and is commonly done on a
recreational basis. Studies examining the effects of smoking cannabis for its analgesic effects
vary in the duration the cannabis was smoked for, the frequency cigarettes were smoked, and
the THC content of the cannabis cigarettes. The range of THC content between studies varied
significantly from 1.29% THC content to 9.4% THC content (27-34). While studies measured
THC content contained in the cigarettes, the content other cannabinoids such as cannabinidiol
were not recorded.
    Smoked cannabis was studied for its efficacy on pain as a result of unspecified neuropathy
(33, 34), HIV induced neuropathy (27, 30), post-traumatic neuropathy (32), multiple sclerosis
(29), and capsaicin injection (31). Of the eight studies on smoked cannabis and pain, all eight
reported a significant decrease in pain (27-34). Among the studies that reported on the clinical
176                            Jonathan K Hwang and Hance Clarke
relevance of pain reductions, there was considerable variation in findings. Wilsey and
colleagues found that 26% of its population on the active agent achieved a clinically relevant
decrease in pain (33), while Abrams and colleagues found that 52% of its population on the
active agent achieved a clinically relevant decrease in pain (27). In addition, one of the studies
found that cannabis’ analgesic effects were dose dependent, reducing pain with 4% THC, but
inducing increased pain at 8% THC (31). Studies generally reported that the side effects of
smoking cannabis were well tolerated, with relatively few serious adverse events (27-29, 31-
34).
     There were no studies that documented the long-term effects of using smoked cannabis as
an analgesic. Long term use of smoked cannabis may result in adverse respiratory effects
similar to those of smoking tobacco (35). More research needs to be done in terms of examining
the long-term tolerability of smoking cannabis and its psychoactive effects at higher cumulative
doses (34).
     As such, smoking cannabis was consistently found to have a statistically significant and
clinically relevant effect in reducing neuropathic pain. As its effects on other types of pain have
yet to be studied, more research is needed to determine its effects. The vast majority of adverse
events experienced were only mild to moderate in severity and generally people tolerated
smoking cannabis quite well. A major limitation of studies examining the effect of smoking
cannabis is that the blinding is often compromised, as patients are often able to differentiate
between smoking the active agent versus placebo (27, 29-31). The potential loss of blinding
should be taken into account when examining the positive effects of smoking cannabis on pain.
                                       CONCLUSION
More research is needed in order to better determine the efficacy of cannabis as an analgesic
treatment. Although the results generally show that cannabis does have a statistically significant
positive effect on pain, the quality of the literature needs improvement. Despite the large
proportion of RCTs in the cannabis and pain literature, many studies have small sample sizes
and possible conflicts of interest that could potentially bias results. Of the three methods of
administration, smoking cannabis had the fewest severe adverse effects and had the largest
number of studies favouring its effects on pain. Nabiximol sprays also had a large number of
studies that favoured its effects on pain, however the clinical significance of the effect varied
between studies. Although adverse events were generally mild to moderate for nabiximols, a
relatively large number of severe adverse events did occur in a single study. The data on
dronabinol capsules was far more mixed, with some studies showing benefits for chronic pain
and other studies showing no analgesic effects. Nabilone had a reasonably large body of
evidence favouring its effect on pain, however it’s clear that more research is needed on
cannabis extract capsules to verify their efficacy. The body of literature on cannabis and pain
as a whole is small and of variable quality. In addition, the majority of studies are very small
RCTs. Larger RCTs would help further confirm the findings presented in the existing data and
would lend credibility to the existing treatments.
                                          Cannabis and pain                                                177
                                      ACKNOWLEDGMENTS
Dr. Clarke is supported by a Merit Award from the Department of Anesthesia at the University
of Toronto.
                                             REFERENCES
[1]    Killestein J, Hoogervorst EL, Reif M, Kalkers NF, Van Loenen AC, Staats PG, et al. Safety, tolerability,
       and efficacy of orally administered cannabinoids in MS. Neurology 2002; 58(9): 1404-7.
[2]    Nurmikko TJ, Serpell MG, Hoggart B, Toomey PJ, Morlion BJ, Haines D. Sativex successfully treats
       neuropathic pain characterised by allodynia: a randomised, double-blind, placebo-controlled clinical
       trial. Pain 2007; 133(1-3): 210-20.
[3]    Berman JS, Symonds C, Birch R. Efficacy of two cannabis based medicinal extracts for relief of central
       neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Pain 2004;
       112(3): 299-306.
[4]    Svendsen KB, Jensen TS, Bach FW. Does the cannabinoid dronabinol reduce central pain in multiple
       sclerosis? Randomised double blind placebo controlled crossover trial. BMJ 2004; 329(7460): 253.
[5]    Wade DT, Robson P, House H, Makela P, Aram J. A preliminary controlled study to determine whether
       whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clin Rehabil 2003;
       17(1): 21-9.
[6]    Hoggart B, Ratcliffe S, Ehler E, Simpson KH, Hovorka J, Lejcko J, et al. A multicentre, open-label,
       follow-on study to assess the long-term maintenance of effect, tolerance and safety of THC/CBD
       oromucosal spray in the management of neuropathic pain. J Neurol 2015; 262(1): 27-40.
[7]    Johnson JR, Lossignol D, Burnell-Nugent M, Fallon MT. An open-label extension study to investigate
       the long-term safety and tolerability of THC/CBD oromucosal spray and oromucosal THC spray in
       patients with terminal cancer-related pain refractory to strong opioid analgesics. J Pain Symp Manage
       2013; 46(2): 207-18.
[8]    Haroutiunian S, Rosen G, Shouval R, Davidson E. Open-label, add-on study of tetrahydrocannabinol
       for chronic nonmalignant pain. J Pain Palliat Care Pharmacother 2008; 22(3): 213-7.
[9]    Rog DJ, Nurmikko TJ, Young CA. Oromucosal delta9-tetrahydrocannabinol/cannabidiol for
       neuropathic pain associated with multiple sclerosis: an uncontrolled, open-label, 2-year extension trial.
       Clin Ther 2007; 29(9): 2068-79.
[10]   Bestard JA, Toth CC. An open-label comparison of nabilone and gabapentin as adjuvant therapy or
       monotherapy in the management of neuropathic pain in patients with peripheral neuropathy. Pain
       Practice 2011; 11(4): 353-68.
[11]   Rintala DH, Fiess RN, Tan G, Holmes SA, Bruel BM. Effect of dronabinol on central neuropathic pain
       after spinal cord injury: a pilot study. Am J Phys Med Rehab 2010; 89(10): 840-8.
[12]   Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R, Fallon MT. Multicenter,
       double-blind, randomized, placebo-controlled, parallel-group study of the efficacy, safety, and
       tolerability of THC: CBD extract and THC extract in patients with intractable cancer-related pain. J Pain
       Symp Manage 2010; 39(2): 167-79.
[13]   Lynch ME, Cesar-Rittenberg P, Hohmann AG. A double-blind, placebo-controlled, crossover pilot trial
       with extension using an oral mucosal cannabinoid extract for treatment of chemotherapy-induced
       neuropathic pain. J Pain Symp Manage 2014; 47(1): 166-73.
[14]   Portenoy RK, Ganae-Motan ED, Allende S, Yanagihara R, Shaiova L, Weinstein S, et al. Nabiximols
       for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-
       controlled, graded-dose trial. J Pain 2012; 13(5): 438-49.
[15]   Rog DJ, Nurmikko TJ, Friede T, Young CA. Randomized, controlled trial of cannabis-based medicine
       in central pain in multiple sclerosis. Neurology 2005; 65(6): 812-9.
178                                Jonathan K Hwang and Hance Clarke
[16]   Langford RM, Mares J, Novotna A, Vachova M, Novakova I, Notcutt W, et al. A double-blind,
       randomized, placebo-controlled, parallel-group study of THC/CBD oromucosal spray in combination
       with the existing treatment regimen, in the relief of central neuropathic pain in patients with multiple
       sclerosis. J Neurol 2013; 260(4): 984-97.
[17]   Kraft B, Frickey NA, Kaufmann RM, Reif M, Frey R, Gustorff B, et al. Lack of analgesia by oral
       standardized cannabis extract on acute       inflammatory pain and hyperalgesia in volunteers.
       Anesthesiology 2008; 109(1): 101-10.
[18]   Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, Group MR. Multiple sclerosis and extract of
       cannabis: results of the MUSEC trial. J Neurol Neurosurg Psychiatry 2012; 83(11): 1125-32.
[19]   Narang S, Gibson D, Wasan AD, Ross EL, Michna E, Nedeljkovic SS, et al. Efficacy of dronabinol
       as an adjuvant treatment for chronic pain patients on opioid therapy. J Pain 2008; 9(3): 254-64.
[20]   Frank B, Serpell MG, Hughes J, Matthews JN, Kapur D. Comparison of analgesic effects and patient
       tolerability of nabilone and dihydrocodeine for chronic neuropathic pain: randomised, crossover, double
       blind study. BMJ 2008; 336(7637): 199-201.
[21]   Toth C, Mawani S, Brady S, Chan C, Liu C, Mehina E, et al. An enriched-enrolment, randomized
       withdrawal, flexible-dose, double-blind, placebo-controlled, parallel assignment efficacy study of
       nabilone as adjuvant in the treatment of diabetic peripheral neuropathic pain. Pain 2012; 153(10): 2073-
       82.
[22]   Turcotte D, Doupe M, Torabi M, Gomori A, Ethans K, Esfahani F, et al. Nabilone as an adjunctive to
       gabapentin for multiple sclerosis-induced neuropathic pain: a randomized controlled trial. Pain Med
       2015; 16(1): 149-59.
[23]   Kalliomaki J, Philipp A, Baxendale J, Annas P, Karlsten R, Segerdahl M. Lack of effect of central
       nervous system-active doses of nabilone on capsaicin-induced pain and hyperalgesia. Clin Exp
       Pharmacol Physiol 2012; 39(4): 336-42.
[24]   Skrabek RQ, Galimova L, Ethans K, Perry D. Nabilone for the treatment of pain in fibromyalgia. J Pain.
       2008; 9(2): 164-73.
[25]   Wissel J, Haydn T, Muller J, Brenneis C, Berger T, Poewe W, et al. Low dose treatment with the
       synthetic cannabinoid Nabilone significantly reduces spasticity-related pain: a double-blind placebo-
       controlled cross-over trial. J Neurol 2006; 253(10): 1337-41.
[26]   Issa MA, Narang S, Jamison RN, Michna E, Edwards RR, Penetar DM, et              al.    The     subjective
       psychoactive effects of oral dronabinol studied in a randomized, controlled crossover clinical trial for
       pain. Clin J Pain 2014; 30(6): 472-8.
[27]   Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful HIV-associated
       sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007; 68(7): 515-21.
[28]   Cooper ZD, Comer SD, Haney M. Comparison of the analgesic effects of dronabinol and smoked
       marijuana in daily marijuana smokers. Neuropsychopharmacology 2013; 38(10): 1984-92.
[29]   Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H, et al. Smoked cannabis for
       spasticity in multiple sclerosis: a randomized, placebo-controlled trial. CMAJ Canadian Medical
       Association Journal. 2012; 184(10): 1143-50.
[30]   Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, et al. Smoked medicinal
       cannabis for neuropathic pain in HIV: a randomized,               crossover          clinical        trial.
       Neuropsychopharmacology. 2009; 34(3): 672-80.
[31]   Wallace M, Schulteis G, Atkinson JH, Wolfson T, Lazzaretto D, Bentley H, et al. Dose-dependent
       effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers.
       Anesthesiology 2007; 107(5): 785-96.
[32]   Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, et al. Smoked cannabis for chronic
       neuropathic pain: a randomized controlled trial. CMAJ 2010; 182(14): e694-701.
[33]   Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Donaghe H. Low-dose vaporized cannabis
       significantly improves neuropathic pain. J Pain 2013; 14(2): 136-48.
[34]   Wilsey B, Marcotte T, Tsodikov A, Millman J, Bentley H, Gouaux B, et al. A randomized, placebo-
       controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain 2008; 9(6): 506-21.
[35]   Martinasek MP, McGrogan JB, Maysonet A. A systematic review of the respiratory effects of
       inhalational marijuana. Respir Care 2016 [Epub ahead of print].
In: Cannabis: Medical Aspects                                            ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                   © 2017 Nova Science Publishers, Inc.
Chapter 14
      The use of medical cannabis for pain in adolescence is controversial. The purpose of this
      chapter is to summarize the literature regarding efficacy, adverse bio-psychosocial and
      neurodevelopmental effects, as well as legislation and utilization patterns of adolescents.
      Clinical trials demonstrating efficacy of cannabis for adolescent pain are essentially
      nonexistent. Adolescent marijuana use may result in a number of physical symptoms, has
      a strong association with schizophrenia, may affect neurocognitive development in the not
      fully mature adolescent brain, and may lead to school, relationship and work problems as
      well as motor vehicle accidents. The proportion of legal medical cannabis patients under
      the age of 18 years ranges from 0.04% to 0.36% in different US states with medical
      marijuana laws. Leading reasons for its being given are pain and epilepsy. Adolescents
      who use medical cannabis have an increased likelihood of displaying high-risk drug related
      behavior. Diversion of medical cannabis to other teenagers is not uncommon. There is a
      need for clinical trials of cannabis for pain control in different adolescent health conditions.
      Clinical guidelines should be prepared to aid physicians and legislators in determining
      when medical marijuana is justified in this age group, and in identifying adolescents likely
      to misuse the drug.
                                          INTRODUCTION
The use of cannabis, referring to drugs derived from the genus cannabis, or its synthetic
cannabinoid derivatives, for treating disease or alleviating symptoms has increased
dramatically in recent years. Chronic or severe pain is one of the most common, if not the
leading, indication for its use.
    Medical cannabis for adolescent pain is controversial. This is because of the scant
knowledge base regarding efficacy, and because of marijuana’s untoward effects – making
decisions for this age group more difficult than for the adult population. The purpose of this
*
    Correspondence: Barry Knishkowy, MD, MPH, POBox 80716, Mevasseret Zion, 90805, Israel. E-mail:
      knishkowyb@gmail.com.
180                                     Barry Knishkowy
review is to summarize the literature regarding clinical trials, adverse bio-psychosocial and
neurodevelopmental effects, as well as legislation and utilization patterns of adolescents.
Suggestions for future research and policy development are offered.
In the United States, there were 1,246,170 legal medical cannabis patients in states with medical
marijuana laws, or an average of 8.06 patients per 1,000 residents in March 2016 (19). Only a
few states provide the age distribution of medical marijuana registered patients. There are data
regarding the number and percentage of patients under the age of 18 years available for Arizona
(42 patients, 0.15%), Colorado (343 patients, 0.32%; 215 under the age of 11), Montana (6
patients, 0.04%), Nevada (41 patients, 0.26%) and Oregon (278 patients, 0.36%; 95 under the
age of 11) (20-24).
     In most states, severe or chronic pain is by far the most common condition for receiving
medical marijuana (13). Data from Colorado show that for children and adolescents in the 0-
10 and 11-17 year age groups, the leading reason is ‘seizures’, whereas it is ‘severe pain’ for
older age groups (21). In Oregon, for children and adolescents under the age of 18, the leading
reasons for receiving medical cannabis are: severe pain (48.6%), seizures (36.0%), spasms
(21.6%), and nausea (17.6%) (24).
     Differences in the rates of use and the diagnoses of registered patients among the states
may result not only from different population characteristics, but from different regulations in
the various medical marijuana programs. There is considerable variability in features of the
various policies, with regard to the number (ranging from 6 to 40) and types of conditions for
which medical marijuana may be given (25). Thus, adolescents with chronic pain from a
specific illness may have legal access to marijuana in one state but not in another.
Several surveys of medical cannabis users have been conducted in different countries/US states,
with the greatest number in the state of California. Various sample selection methods were
used, and the study groups were generally not shown to be representative of the entire medical
cannabis-using population.
     One survey in California, which examined data on 1,746 consecutive admissions to nine
medical marijuana clinics in 2006, included data on adolescents. Of the population surveyed,
0.2% were 12-18 years old, and 17.8% were 18-24 years old. For the entire study population,
82.6% used the marijuana for pain relief, 70.6% for sleep, 55.6% for relaxation, 41.3% for
spasms, and 40.8% for headache. The most common physician diagnoses were low back pain
(26.2%), anxiety disorders (18.7%) and arthritis (18.0%). Of note, 50.8% used the marijuana
as a substitute for prescription medication, often opioids (26).
In the United States, three periodic surveys of the adolescent population track substance use,
including marijuana: Monitoring the Future (MTF), the Centers for Disease Control and
Prevention’s Youth Risk Behavior Survey (YRBS), and the National Survey on Drug Use and
Health (NSDUH) (10).
     Monitoring the Future provided the first nationally representative sample of 12th graders’
medical marijuana use. Data from the 2012 and 2013 MTF study were analyzed to determine
184                                      Barry Knishkowy
the percentage of teens who were illicit users (source of marijuana was illegal or nonmedical),
medical users, or diverted medical marijuana users (received their marijuana from someone
else’s marijuana prescription) during the past year. In this sample of 4,394 adolescents, 28.8%
were illicit users, 1.1% were medical users, and 6.1% were diverted medical marijuana users.
Compared with illicit users, medical users had a higher odds of engaging in other high-risk
behaviors (such as using illegal prescription drugs or illicit drugs other than marijuana), while
those using diverted marijuana had a higher odds of engaging in all of the other behaviors
studied (27).
First and foremost, the use of medical cannabis by adolescents is currently limited because of
a lack of data demonstrating efficacy and the high potential for current and future untoward
health effects in this age group. These are among the reasons for policy statements by several
professional organizations opposing medical marijuana use in this population. The American
Academy of Pediatrics, for example, is against cannabinoid administration to children and
adolescents except for FDA-approved indications, and possibly for patients with life-limiting
or severely debilitating illnesses for whom current therapies are inadequate (7).
     Additional considerations may play into decisions regarding adolescent use of medical
cannabis in general, and for pain in particular. Data from the MTF survey show that high-risk
drug-related behavior was more prevalent among medical marijuana users than among illicit
marijuana users (27). Similar findings were reported in the study of adolescent and young adult
cancer patients cited previously, where 11.7% of 94 patients given opioid therapy displayed
aberrant opioid-associated behavior (AOB). In that study, psychosocial risk factors were
identified in 90.9% of those with AOB, with a past or current mental health diagnosis probably
being the most significant risk factor for opioid abuse (3). Data comparing adolescent and
young adult non-cancer patients with older adults have shown that the younger patients are
more likely to abuse opioids and illicit substances (28). These studies raise the question of
which adolescent medical cannabis users might be at relatively high risk for aberrant drug-
related behavior.
     In contrast, some studies (mainly involving adults) have shown that medical cannabis use
is associated with a decrease or cessation in the use of opioids and other medications for pain
control (26, 29). And so, if one views cannabis use as a less problematic option than opioids, it
may constitute a positive alternative in some cases.
     There are also public health issues related to the legalization and use of medical cannabis.
Several studies have looked at the effect of such legislation upon recreational marijuana use
among youth, with conflicting results (30, 31). Although there is no solid evidence that
adolescent marijuana use increases when medical marijuana is legalized, the phenomenon of
large-scale medical cannabis diversion (27), including to high-risk populations (32), cannot be
ignored.
                            Medical cannabis for pain in adolescence                                    185
                                              SUMMARY
      1. Data regarding the efficacy of cannabis in the treatment of adolescent pain are
         essentially nonexistent.
      2. There are significant health risks entailed in the use of cannabis, many of which are
         unique to children and adolescents.
      3. Adolescents using medical cannabis display high-risk drug related behavior more
         frequently than recreational users.
      4. The utilization rates of legally prescribed medical cannabis among children and
         adolescents are proportionately very low, ranging from 0.04% to 0.36% of all patients
         in published state statistical reports.
      5. Common reasons for using medical marijuana in children and adolescents, based on
         very limited data, are pain control and seizures.
      6. Public health problems, including medical cannabis diversion to other teenagers, may
         result from its legal, medical utilization.
                                            CONCLUSION
The approach to adolescent use of medical cannabis is inherently different than that for adults.
Two major challenges follow from this review of the topic.
    The first is to conduct research regarding which conditions causing significant pain in this
age group respond to treatment with cannabinoids, and when that treatment would be preferred
to other therapeutic options. Further research is also needed to determine risk factors for
potential medical cannabis misuse.
    The second challenge is to develop, with limited current knowledge, clinical guidelines to
help clinicians and legislators decide on the indications and contraindications for medical
marijuana use among youth, and to determine exclusion criteria and risk reduction measures
based upon risk factor assessment.
                                            REFERENCES
[1]     King S, Chambers CT, Huguet A, MacNevin RC, McGrath PJ, Parker L, et al. The epidemiology of
        chronic pain in children and adolescents revisited: a systematic review. Pain 2011;152(12):2729-38.
[2]     Landry BW, Fischer PR, Driscoll SW, Koch KM, Harbeck-Weber C, Mack KJ, et al. Managing chronic
        pain in children and adolescents: a clinical review. PM R 2015;11(Suppl):S295-315.
[3]     Ehrentraut JH, Kern KD, Long SA, An AQ, Faughnan LG, Anghelescu DL. Opioid misuse behaviors
        in adolescents and young adults in a hematology/oncology setting. J Pediatr Psychol 2014;39(10):1149-
        60.
[4]     Hill KP. Medical marijuana for treatment of chronic pain and other medical and psychiatric problems:
        A clinical review. JAMA 2015;313(24):2474-83.
[5]     Rieder MJ; Canadian Paediatric Society Drug Therapy and Hazardous Substances Committee. Is the
        medical use of cannabis a therapeutic option for children? Paediatr Child Health 2016;21(1):31-4.
[6]     Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, et al. Cannabinoids for
        medical use: A systematic review and meta-analysis. JAMA 2015;313(24):2456-73.
186                                            Barry Knishkowy
[7]    Committee on Substance Abuse, Committee on Adolescence. The impact of marijuana policies on
       youth: clinical, research and legal update. Pediatrics 2015;135(3):584-7.
[8]    Greydanus DE, Hawver EK, Greydanus MM, Merrick J. Marijuana: Current concepts. Front Public
       Health 2013;1:42.
[9]    Hadland SE, Harris SK. Youth marijuana use: state of the science for the practicing clinician. Curr Opin
       Pediatr 2014;26(4):420-7.
[10]   Ammerman S, Ryan S, Adelman W; Committee on Substance Abuse, Committee on Adolescence. The
       impact of marijuana policies on youth: clinical, research and legal update. Pediatrics 2015;135(3):e769-
       85.
[11]   Tamm L, Epstein JN, Lisdahl KM, Molina B, Tapert S, Hinshaw SP, et al; MTA Neuroimaging Group.
       Impact of ADHD and cannabis use on executive functioning in young adults. Drug Alcohol Depend
       2013;133(2):607-14.
[12]   Hartman RL, Huestis MA. Cannabis effects on driving skills. Clin Chem 2013;59(3):478-92.
[13]   24 Legal medical marijuana states and DC: laws, fees and possession limits. URL:
       http://medicalmarijuana.procon.org/view.resource.php?resourceID=000881.
[14]   Lucas P. It can’t hurt to ask; a patient-centered quality of service assessment of health canada’s medical
       cannabis policy and program. Harm Reduct J 2012;9:2.
[15]   Legality of cannabis by country. URL: http://en.wikipedia.org/wiki/Legality_of_cannabis_ by_country.
[16]   Marinol capsules. URL: http://www.rxabbvie.com/pdf/marinol_PI.pdf.
[17]   Full prescribing information – Cesamet. URL: https://www.cesamet.com/pdf/Cesamet_PI_50_
       count.pdf.
[18]   GWPharma – Sativex. URL: http://www.gwpharm.com/Sativex.aspx.
[19]   Number of legal medical marijuana patients. URL: http://medicalmarijuana.procon.org/
       view.resource.php?resourceID=005 889.
[20]   Arizona Department Health Services: Medical Marijuana. URL: azdhs.gov/licensing/medical-
       marijuana/index.php.
[21]   Colorado Department of Public Health and Environment: Medical Marijuana Registry. URL:
       https://www.colorado.gov/pacific/cdphe/medicalmarijuana.
[22]   Montana Marijuana Program. URL: https://dphhs.mot.gov/marijuana.
[23]   Nevada Division of Public and Behavioral Health: Medical marijuana. URL: dpbh.nv.gov/Reg/
       Medical_Marijuana/.
[24]   Oregon Medical Marijuana Program (OMMP). URL: public.health.oregon.gov/Diseases Conditions/
       ChronicDisease/MedicalM arijuanaProgram/Pages/index/aspx.
[25]   Bestrashniy J, Winters KC. Variability in medical marijuana laws in the United States. Psychol Addict
       Behav 2015;29(3):639-42.
[26]   Nunberg H, Kilmer B, Pacula RL, Burgdorf J. An analysis of applicants presenting to a medical
       marijuana specialty practice in California. J Drug Policy Anal 2011;4(1):1-16.
[27]   Boyd CJ, Veliz PT, McCabe SE. Adolescents’ use of medical marijuana: a secondary analysis of
       Monitoring the Future data. J Adolesc Health 2015;57(2):241-4.
[28]   Pergolizzi JV Jr, Gharibo C, Passik S, Labhsetwar S, Taylor R Jr, Pergolizzi JS, et al. Dynamic risk
       factors in the misuse of opioid analgesics. J Psychosom Res 2012;72(6);443-51.
[29]   Swift W, Gates P, Dillon P. Survey of Australians using cannabis for medical purposes. Harm Reduct
       J 2005;2:18.
[30]   Choo EK, Benz M, Zaller N, Warren O, Rising KL, McConnell KJ. The impact of state medical
       marijuana legislation on adolescent marijuana use. J Adolesc Health 2014;55(2):160-6.
[31]   Wall MM, Poh E, Cerda M, Keyes KM, Galea S, Hasin DS. Commentary on Harper S, Strumph EC,
       Kaufman JS. Do medical marijuana laws increase marijuana use? Replication study and extension. Ann
       Epidemiol 2012;22(7):536-7.
[32]   Salomonsen-Sautel S, Sakai JT, Thurstone C, Corley R, Hopfer C. Medical marijuana use among
       adolescents in substance abuse treatment. J Am Acad Child Adolesc Psychiatry 2012;51(7):694-702.
SECTION FOUR: POLICY, ETHICS AND SOCIAL
             COMMENTARY
In: Cannabis: Medical Aspects                                                 ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                        © 2017 Nova Science Publishers, Inc.
Chapter 15
            In 2013 the Canadian Government enacted the Marihuana for Medical Purposes
       Regulations, allowing licensed marihuana producers to grow and sell medical marihuana
       to patients for a variety of medical conditions, including pain. Patients require a medical
       document from a health professional in order to register with a licensed producer, so
       physicians are now being asked to provide a prescription for a drug that has not undergone
       standardized Health Canada regulations and without clear dosing guidelines. The focus of
       this chapter is to review the legislation, the Health Canada document on Medical
       Marihuana and the Canadian Pain Society consensus statement on the management of
       neuropathic pain in order to highlight the role of marihuana in the management of
       neuropathic pain and propose a prescribing strategy for medical marihuana.
                                             INTRODUCTION
In 2001 the Canadian government permitted possession and use of marihuana for medical
purposes via the Marihuana Medical Access Regulation (MMAR) (1). Patients were required
to obtain authorization from Health Canada, subsequent to validation from a physician that they
suffered from a condition for which medical marihuana use was approved. In 2013, in order to
address concerns about the lack of standards in the production of marihuana by small producers
and the cumbersome authorization process, the government replaced the MMAR with the
Marihuana for Medical Purposes Regulations (MMPR) (2, 3).
*
    Corresponding author: Hance Clarke, MD, Staff Anesthesiologist, Director of Clinical Pain Services and Medical
       Director of Pain Research Unit, Toronto General Hospital, 200 Elizabeth Street, Toronto, ON Canada. E-mail:
       Hance.Clarke@uhn.ca.
190                  Ainsley M Sutherland, Judith Nicholls and Hance Clarke
     Under the MMPR producers were granted licenses to grow and distribute dried marihuana
for medical purposes. Production practices, packaging and labelling, security measures and
transportation are all now closely regulated under the MMPR. Licensed producers must report
to provincial licensing authorities (2, 3). Patients no longer have to apply to Health Canada for
authorization to possess marihuana. They can now ask their physician for a medical document
for medical marihuana, to be filled by one of the licensed producers (2, 3). Licensed producers
can provide dried marihuana to another licensed producer, a licensed dealer, the Health
Minister, or a person (patient) who is registered with the licensed producer after submitting a
medical document confirming their eligibility for medical marihuana. The medical document
must specify how many grams of marihuana per day the patient is permitted, which is akin to
a prescription.
     This poses the question then: how do we as physicians prescribe marihuana? In our practice
there has been an exponential increase in the number of patients requesting prescriptions for
medical marihuana for chronic pain. There are, however, no large randomized controlled trials
examining the efficacy of medical marihuana in various pain syndromes. A number of small
trials in patients suffering from painful diabetic neuropathy, HIV neuropathy and post-
traumatic/post-surgical neuropathic pain have shown medical marihuana to be more efficacious
than placebo for pain control (4-9). However, these trials need to be replicated with larger
numbers of patients. There are no dosing guidelines, and it remains unclear what doses patients
receive when they inhale medical marihuana. Furthermore, we remain unclear as to which
component of medical marihuana is responsible for the analgesic effects as Cannabis plants
contain at least 489 distinct compounds from 18 different chemical classes (10).
     For these reasons Health Canada clearly states on the front page of its document –
Cannabis: Information for Health Care Professionals that “Cannabis is not an approved
therapeutic product and the provision of this information should not be interpreted as an
endorsement of the use of this product, or cannabis generally, by Health Canada” (11).
However, physicians are expected to provide prescriptions without high quality evidence of
efficacy, or dosing guidelines.
     In this chapter, we will summarize Health Canada’s Information for Health Care
Professionals on Cannabis as a starting point for the rational prescription of medical marihuana
in Canada. Many more high-quality randomized controlled trials and dose-finding trials will be
required before we can establish a true evidenced based approach to prescribing marihuana for
pain.
                                      PATIENT CASE
A 62-year-old woman is referred to a tertiary care chronic pain clinic with a two-year history
of debilitating neuropathic pain following a left-sided mastectomy for breast cancer. Her pain
has led to severe functional decline. She reports that she has constant burning pain and
paresthesias on the left lateral chest, and her clothes cause her severe pain when they brush
against the skin. Her mood is low, and she is not sleeping because of the pain. She rarely leaves
her house now except for medical appointments. She is desperate for help with her pain, and
her daughter suggested trying medical marihuana. Her family doctor is not comfortable
                 Medical cannabis from the pain physician’s perspective                      191
prescribing medical marihuana, and so has referred her to the pain clinic. How should this
patient be counselled and managed?
The Canadian Pain Society released a revised consensus statement in 2014 regarding the
pharmacological management of chronic neuropathic pain (12). The guideline stresses the
importance of assessing all patients with chronic neuropathic pain for comorbidities such as
anxiety and depression, and to focus the goals of treatment on improvement in function and
quality of life (12). It is important to keep these goals in mind when considering the initiation
of therapy with medical marihuana. First and second line agents should be considered before
trialing medical marihuana.
     First-line agents for neuropathic pain are the anticonvulsants gabapentin and pregabalin,
tricyclic antidepressants (TCAs) and the serotonin-norepinephrine reuptake inhibitors (SNRIs)
duloxetine and venlafaxine (12). Patients should have an adequate trial of these medications,
preferably in combination, before other modalities such as cannabinoids are considered.
     Second-line agents include the weak opioid agonist tramadol and other opioid analgesics
(12). The number needed to treat (NNT) for opioids in neuropathic pain syndromes is 2.6 (13).
With the use of opioids, however, the risk of adverse events increases significantly, including
respiratory depression, drowsiness, and the risk of abuse and addiction. Many older patients
with medical comorbidities do not tolerate treatment with opioids. In patients with several
complex co-morbidities (e.g., renal failure, cardiac compromise, encephalopathy) it may be
justified to consider cannabinoids before opioids, or as adjuncts to reduce opioid requirements,
because initiating opioids in these patients may lead to sudden decline.
     The major change in the Canadian Pain Society Consensus Statement in 2014 from the
previous statement in 2007 was to list cannabinoids as third-line agents in the management of
neuropathic pain. In the 2007 statement, they were listed in as fourth-line agents (12). This
change was based on a systematic review of trials of cannabinoids in chronic pain that found
there had been six positive studies out of seven high quality studies investigating cannabinoids
in chronic neuropathic pain (14). These studies, however, were all small and examined the
efficacy of different cannabinoids in different pain syndromes. Although the results of these
studies are intriguing, they are inadequate for evidence-based practice.
     Fourth line agents now include selective serotonin reuptake inhibitors (SSRIs), other
anticonvulsants such as lamotrigine, lacosamide, topiramate and valproic acid, methadone and
topical lidocaine (12).
THC), Δ8-THC, and cannabinol (CBN) from cannabis sitava, or marihuana. Δ9-THC is
responsible for the psychoactive effects of cannabis (15). Cannabidiol (CBD) is a component
of cannabis that is thought to possess anti-inflammatory and analgesic properties, but it does
not act through the CB1/2 receptors at physiological concentrations (16) and has minimal
psychotropic effects. Data does not exist regarding the ideal ratio of THC to CBD for treating
pain.
     Activation of the CB1 or CB2 receptors by endo- or phytocannabinoids results in
inhibition of adenylyl cyclase activity, and decreased formation of cyclic AMP (17) leading
to among other things, inhibition of neurotransmitter release in the CNS (17,18). This
includes neurotransmitters with important roles in pain transmission and modulation (5-
hydroxytryptamine (5-HT), glutamate, acetylcholine, γ-Aminobutyric acid (GABA), and
noradrenaline) (18). CB1 and CB2 receptors are expressed throughout the body and brain (19),
this presents a challenge in using phytocannabinoids for pain without adverse effects such as
cognitive dysfunction.
     The regular/recreational cannabis user is a very unique clinical situation. The surge in
registered patents to date is likely linked to many recreational users seeking their licenses for
various therapeutic causes such as insomnia, anxiety, nausea, insomnia, or pain. The question
becomes how to rationally and safely prescribe medical marihuana given many long time users
consume strains with very high THC content (e.g., >25%) and the ethics behind this practice.
Several patients in our clinic have been victims of assault and there is a harm reduction
component for individuals that will continue to consume recreational cannabis sought outside
of the regulated licensed producers. According to the MMPR the health care professional must
indicate the daily quantity of dried marihuana to be used by the person, expressed in grams (2).
But what is an appropriate daily amount for a patient with pain? And what dose of
phytocannabinoids with analgesic properties will the patient actually receive by inhaling dried
marihuana?
                                                                          Serum [Δ9-THC]
   Cannabinoid     Components          Dose         Onset    Duration
                                                                              (range)
   Nabilone        Δ9-THC          2mg BID         60-       8-12h       2ng/mL
                                                   90min
   Dronabinol      Δ9-THC          2.5mg BID       30-       4-6h        1.3ng/mL (0.7 -
                                   5mg BID         60min                 1.9ng/mL)
                                   10mg BID                              2.9ng/mL (1.2 –
                                                                         4.7ng/mL)
                                                                         7.9ng/mL (3.3 –
                                                                         12.4ng/mL)
   Nabiximols      Δ9-THC +        4 sprays        15-       2-4h        5.5ng/mL
                   CBD             (10.8mg Δ9-     40min                 CBD: 3ng/mL
                                   THC and
                                   10mg CBD)
   Dried                           1.75% (16mg     5min      2-4h        7.0 ± 8.1ng/mL
   Marihuana                       Δ9-THC)
                                   3.55% (34mg                           18.1 ± 12.0ng/mL
                                   Δ9-THC)
to remember that the smoked dose is the amount of Δ9-THC in milligrams that is available in
the entire marihuana cigarette. The actual amount of Δ9-THC delivered to the patient is highly
variable as it is dependent on smoking technique, effort, number of puffs, etc.
     Keeping in mind the variability in the dose actually received by the patient due to
variability in inhalation, a reasonable smoked dose for analgesia may be around 30-150mg per
day of THC. A study of smoked cannabis in neuropathic pain found patient’s visual analogue
scores (VAS) decreased after inhalation of the equivalent of either 28mg or 56mg per day of
THC compared to placebo, but there was no difference between the lower and higher doses (5).
Another study in chronic post-surgical or post-traumatic pain found that patients receiving only
the equivalent of 7mg per day of THC reduced pain and improved sleep (7). A smoked dose of
7mg a day appears to be very low however, in light of a titration study in HIV neuropathy that
found most patients self-titrated to a smoked dose between 64-128mg THC per day (6). Only
one patient out of 28 in this trial titrated to a lower smoked dose of 32mg per day of THC (6).
Another study in HIV neuropathy found patients had decreased pain with a smoked dose of
96mg per day of THC versus placebo (4). This is in line with the finding that in patients on
chronic opioids for a variety of pain conditions, a smoked dose of 96mg/day of THC decreased
their pain scores by 27% with no change in the pharmacokinetics of the opioids (26). Based on
these studies it would be reasonable to recommend a patient with a neuropathic pain condition
begin with a smoked dose of 30mg of THC per day and titrate up or down according to the
degree of analgesia they achieve while avoiding side effects and cognitive dysfunction.
     Surveys of medical marihuana patients show that the average consumption is 1-3g of dried
marihuana per day (27-29). A reasonable starting prescription would be around 1-2g per day
recognizing that content of Δ9-THC, and thus the smoked dose, in any given gram of dried
marihuana is highly variable. According to Health Canada the average THC content in illicit
marihuana is 10% (11). The dried marihuana provided by Health Canada has a THC content of
12.5  2% and less than 0.5% CBD (11). Licensed producers have a wide range of products
with varying THC content (0.5-27% in one case), and CBD (0-18% in one case) (30). Cannabis
naïve patients being initiated on medical marihuana therapy should be advised to start
with low THC (i.e., less than 10-15%) content to avoid adverse psychotropic effects.
Several cannabis naïve patents in our clinic have presented to emergency departments having
acquired marihuana from dispensaries and licensed producers with THC content greater than
20% THC experiencing intolerable hallucinogenic effects of the strain. Patients without any
prior experience need to be warned of this potential. Licensed producers list the THC and CBD
percentages on their websites, so the patient can select a product with a low THC content.
Patients in consultation with their medical practitioners can then titrate the amount of
marihuana smoked, the THC and CBD content, to achieve a therapeutic effect with minimal
adverse effects.
     Currently medical marihuana is not legal for oral consumption in edible form under the
MMRP. There exists no solid evidence for converting a smoked dose to an oral dose of
marihuana, however, Health Canada suggests using a conversion factor of 2.5 to account for
differences in bioavailability between smoking (25% bioavailability) and orally ingesting (10%
bioavailability) marihuana (11,31). Thus, if a patient has good therapeutic effect from smoking
1g of medical marihuana with 10% THC (0.1 x 1g = 100mg THC), they may need to ingest
2.5g of the same strain with 10% THC (250mg THC) to achieve the same therapeutic effect
(smoked dose (mg) x 2.5 = oral dose (mg)) (see Table 2) (11). Health Canada provides a table
for quick conversion from smoke to oral dose of marihuana (see Table 3) (11). As stated above,
                 Medical cannabis from the pain physician’s perspective                   195
patients should be advised that marihuana in any form must be heated above 120°C in order to
decarboxylate inactive THCA to the active form (THC) (32).
                “Smoked dose”
     % THC in a 750 mg cannabis cigarette           Estimated oral dose (mg Δ9-THC)
         (Total available mg Δ9-THC)
   1% THC (7.5 mg)                                                18.8 mg
   2% THC (15 mg)                                                 37.5 mg
   2.5% THC (18.8 mg)                                             46.8 mg
   3% THC (22.5 mg)                                               56.3 mg
   5% THC (37.5 mg)                                               93.8 mg
   7.5% THC (56.3 mg)                                            140.6 mg
   10% THC (75 mg)                                               187.5 mg
   12.5% THC (93.8 mg)                                           234.4 mg
   15% THC (112.5 mg)                                            281.3 mg
   20% THC (150 mg)                                               375 mg
    Medical marihuana may also be used as an adjunct to wean opioids. In one such case a
patient with chronic abdominal pain was weaned from an equivalent of hydromorphone 30mg
196                   Ainsley M Sutherland, Judith Nicholls and Hance Clarke
per day following a liver transplant to a total daily dose of 6mg per day with the addition of
medical marihuana (39). This patient used 1g of high CBD:low THC cannabis strain per day
for analgesia (THC 0.79%, CBD 17.08%) (39). Another study found that the use of the
cannabinoid dronabinol in patients undergoing detoxification from opioids was associated with
a decrease in the severity of withdrawal symptoms and that patients who smoked marihuana
were more likely to complete detoxification (40).
                                          CONCLUSION
Neuropathic pain is one of the most difficult conditions to treat effectively, and the evidence
for any of the therapies currently recommended is weak. A step-wise approach to treatment
based on the 2014 Canadian Pain Society consensus statement should be used for all patients.
Cannabinoids are considered third-line treatment and should only be considered when other
treatment modalities have failed, or to supplement these modalities in cannabinoid naïve
patients. For the cannabinoid naïve patient, we suggest trying oral synthetic cannabinoids such
as nabilone first. If this therapy is not successful, or if the patient is already using medical
marihuana, start patients on a low smoked dose of around 30mg of THC and titrate to analgesia
and side effects.
     Early clinical trials are promising, but they are small with varying study designs and pain
disorders. With the legalization of medical marihuana in Canada and the trend towards
legalization in the United States there is hope for better data and clearer, evidence-based
prescribing guidelines in the future. The most helpful evidence will come from companies
invested in generating pharmacokinetic data in humans with the aim to generate an oral tablet
that could be reliably absorbed and prescribed.
                                          REFERENCES
[1]   Government of Canada. Marihuana Medical Access Regulations, SOR/2001-227. Government of
      Canada Department of Justice, 2001. URL: http://laws-lois.justice.gc.ca/PDF/SOR-2001-227.pdf.
[2]   Government of Canada: Marihuana for Medical Purposes Regulations, SOR/2013-119. Government of
      Canada Department of Justice 2013. URL: http://www.laws-lois.justice.gc.ca/PDF/SOR-2013-119.pdf.
[3]   Beaulieu P, Boulanger A, Desroches J, Clark AJ. Medical cannabis: considerations for the
      anesthesiologist and pain physician. Can J Anaesth 2016; 63(5): 608-24.
[4]   Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful HIV-associated
      sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007; 68(7): 515-21.
[5]   Wilsey B, Marcotte T, Tsodikov A, Millman J, Bentley H, Gouaux B, et al. A randomized, placebo-
      controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain 2008; 9(6): 506-21.
[6]   Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, et al. Smoked medicinal
      cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology
      2009; 34(3): 672-80.
[7]   Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, et al. Smoked cannabis for chronic
      neuropathic pain: a randomized controlled trial. CMAJ 2010; 182(14): e694-701.
[8]   Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Donaghe H. Low-dose vaporized cannabis
      significantly improves neuropathic pain. J Pain 2013; 14(2): 136-48.
[9]   Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful
      diabetic neuropathy. J Pain 2015; 16(7): 616-27.
                   Medical cannabis from the pain physician’s perspective                              197
[10]   Elsohly MA, Slade D. Chemical constituents of marijuana: the complex mixture of natural cannabinoids.
       Life Sci 2005; 78(5): 539-48.
[11]   Abramovici H. Information for health care professionals: cannabis (marihuana, marijuana) and the
       cannabinoids. Health Canada 2013. URL: http://www.hc-sc.gc.ca/dhp-mps/alt_formats/pdf/
       marihuana/med/infoprof-eng.pdf.
[12]   Moulin D, Boulanger A, Clark AJ, Clarke H, Dao T, Finley GA, et al. Pharmacological management of
       chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Pain Res Manag
       2014; 19(6): 328-35.
[13]   Finnerup NB, Sindrup SH, Jensen TS. The evidence for pharmacological treatment of neuropathic pain.
       Pain 2010; 150(3): 573-81.
[14]   Lynch ME, Campbell F. Cannabinoids for treatment of chronic non-cancer pain; a systematic review of
       randomized trials. Br J Clin Pharmacol 2011; 72(5): 735-44.
[15]   Hajos N, Ledent C, Freund TF. Novel cannabinoid-sensitive receptor mediates inhibition of
       glutamatergic synaptic transmission in the hippocampus. Neuroscience 2001; 106(1): 1-4.
[16]   Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R. Non-psychotropic plant cannabinoids:
       new therapeutic opportunities from an ancient herb. Trends Pharmacol Sci 2009; 30(10): 515-27.
[17]   De Petrocellis L, Di Marzo V. An introduction to the endocannabinoid system: from the early to the
       latest concepts. Best Pract Res Clin Endocrinol Metab 2009; 23(1): 1-15.
[18]   Serrano A, Parsons LH. Endocannabinoid influence in drug reinforcement, dependence and addiction-
       related behaviors. Pharmacol Ther 2011; 132(3): 215-41.
[19]   Di Marzo V, Piscitelli F, Mechoulam R. Cannabinoids and endocannabinoids in metabolic disorders
       with focus on diabetes. Handb Exp Pharmacol 2011(203): 75-104.
[20]   Karschner EL, Darwin WD, Goodwin RS, Wright S, Huestis MA. Plasma cannabinoid
       pharmacokinetics following controlled oral delta9-tetrahydrocannabinol and oromucosal cannabis
       extract administration. Clin Chem 2011; 57(1): 66-75.
[21]   Schwilke EW, Schwope DM, Karschner EL, Lowe RH, Darwin WD, Kelly DL, et al. Delta9-
       tetrahydrocannabinol (THC), 11-hydroxy-THC, and 11-nor-9-carboxy-THC plasma pharmacokinetics
       during and after continuous high-dose oral THC. Clin Chem 2009; 55(12): 2180-9.
[22]   Meda Pharmaceuticals. Cesamet Product Monograph. Montreal, Quebec: Valeant Canada Limited,
       2013.
[23]   GW Pharmaceuticals. Sativex Product Monograph. Wiltshire, United Kingdom: GW Pharmaceuticals
       Limited, 2010.
[24]   Abbott Products Inc. Marinol Product Monograph. Saint-Laurent, Quebec: Abbott Products
       Incorporated, 2010.
[25]   World Health Organization. Cannabis: a health perspective and research agenda. World Health
       Organization Division of Mental Health and Prevention of Substance Abuse 1997. URL:
       http://apps.who.int/iris/bitstream/10665/63691/1/WHO_MSA_PSA_97. 4.pdf.
[26]   Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL. Cannabinoid-opioid interaction in chronic
       pain. Clin Pharmacol Ther 2011; 90(6): 844-51.
[27]   Carter GT, Weydt P, Kyashna-Tocha M, Abrams DI. Medicinal cannabis: rational guidelines for dosing.
       IDrugs 2004; 7(5): 464-70.
[28]   Clark AJ, Ware MA, Yazer E, Murray TJ, Lynch ME. Patterns of cannabis use among patients with
       multiple sclerosis. Neurology 2004; 62(11): 2098-100.
[29]   Ware MA, Adams H, Guy GW. The medicinal use of cannabis in the UK: results of a nationwide survey.
       Int J Clin Pract 2005; 59(3): 291-5.
[30]   MedReleaf. MedReleaf 2016. URL: http://medreleaf.com/.
[31]   Zuurman L, Ippel AE, Moin E, van Gerven JM. Biomarkers for the effects of cannabis and THC in
       healthy volunteers. Br J Clin Pharmacol 2009; 67(1): 5-21.
[32]   Dussy FE, Hamberg C, Luginbuhl M, Schwerzmann T, Briellmann TA. Isolation of Delta9-THCA-A
       from hemp and analytical aspects concerning the determination of Delta9-THC in cannabis products.
       Forensic Sci Int 2005; 149(1): 3-10.
[33]   Wilson AR, Maher L, Morgan MM. Repeated cannabinoid injections into the rat periaqueductal gray
       enhance subsequent morphine antinociception. Neuropharmacology 2008; 55(7): 1219-25.
198                     Ainsley M Sutherland, Judith Nicholls and Hance Clarke
[34]   Smith PA, Selley DE, Sim-Selley LJ, Welch SP. Low dose combination of morphine and delta9-
       tetrahydrocannabinol circumvents antinociceptive tolerance and apparent desensitization of receptors.
       Eur J Pharmacol 2007; 571(2-3): 129-37.
[35]   National Opioid Use Guideline Group. Canadian guideline for safe and effective use of opioids for
       chronic non-cancer pain. Hamilton, Ontario: McMaster National Pain Centre, 2010.
[36]   Klooker TK, Leliefeld KE, Van Den Wijngaard RM, Boeckxstaens GE. The cannabinoid receptor
       agonist delta-9-tetrahydrocannabinol does not affect visceral sensitivity to rectal distension in healthy
       volunteers and IBS patients. Neurogastroenterol Motil 2011; 23(1): 30-5, e2.
[37]   Lal S, Prasad N, Ryan M, Tangri S, Silverberg MS, Gordon A, et al. Cannabis use amongst patients
       with inflammatory bowel disease. Eur J Gastroenterol Hepatol 2011; 23(10): 891-6.
[38]   Naftali T, Lev LB, Yablecovitch D, Half E, Konikoff FM. Treatment of Crohn’s disease with cannabis:
       an observational study. Isr Med Assoc J 2011; 13(8): 455-8.
[39]   Meng H, Hanlon JG, Katznelson R, Ghanekar A, McGilvray I, Clarke H. The prescription of medical
       cannabis by a transitional pain service to wean a patient with complex pain from opioid use following
       liver transplantation: a case report. Can J Anaesth 2016; 63(3): 307-10.
[40]   Bisaga A, Sullivan MA, Glass A, Mishlen K, Pavlicova M, Haney M, et al. The effects of dronabinol
       during detoxification and the initiation of treatment with extended release naltrexone. Drug Alcohol
       Depend 2015; 154: 38-45.
[41]   Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers 2007; 4(8): 1770-804.
In: Cannabis: Medical Aspects                                             ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                    © 2017 Nova Science Publishers, Inc.
Chapter 16
            In 2014, the Canadian federal Marihuana for Medical Purposes Regulations (MMPR)
       shifted the role of authorizing medical cannabis (MC) from Health Canada to physicians.
       Accordingly, provincial and territorial bodies are issuing physician practice standards to
       provide guidance; however, this does not address how healthcare organizations (HCOs),
       like hospitals, handle the use of MC. Therefore, unless provinces and territories pass MC
       legislation, HCOs must develop institutional policy to control the use of MC. Objective:
       We argue that HCOs should consider five policy issues when considering organizational
       MC policy: 1) locus of administration, 2) permissible modes of administration, 3) clinical
       management, 4) security, and 5) the therapeutic relationship. Additionally, associated
       ethical issues should be taken into consideration; namely, harm reduction and principles
       such as respect for autonomy, non-malificence, reciprocity, justice and the harm principle.
       Discussion: The possible modes of MC administration could include smoking,
       vapourizing, ingesting, and oral synthetic preparations, and each raises unique issues for
       HCOs. Clinical management may include authorizing and monitoring for side-effects and
       overseeing a change in route of administration such as from smoking to ingesting. The
       security element addresses avoiding the abuse, misuse and diversion of MC through
       appropriate storage and establishing guidelines for healthcare provider involvement. The
       therapeutic relationship entails recognition that MC is a permitted medical treatment and
       patients should not be unreasonably and unfairly disadvantaged by institutional policy.
       Conclusions: With an evolving Canadian legal and policy landscape, HCOs managing MC
       use in acute care settings are complex but can be informed by key ethical and policy
       considerations outlined herein.
*
    Corresponding author: Ms Sally Bean, Ethics Centre, Sunnybrook Health Sciences Centre, H263-2075 Bayview
       Avenue, Toronto, Ontario, M4N 3M5, Canada. E-mail: sally.bean@sunnybrook.ca.
200                              Sally Bean and Maxwell J Smith
                                     INTRODUCTION
With an evolving Canadian legal and policy landscape surrounding medical cannabis (MC),
the role of healthcare organizations (HCOs) in managing MC use in acute care settings is unique
and complex. Due to the use of ‘medical cannabis’ in the most recent Canadian federal
legislation, we prefer the nomenclature of medical ‘cannabis’ as opposed to ‘marijuana’ or
‘marihuana’, which is variably used in the Canadian legal context.
     Consider the following illustrative case: an end-stage cancer patient has found successful
symptom relief by smoking MC authorized by his family physician in the community. The
patient is admitted to hospital due to an exacerbation of cancer related symptoms and wishes to
continue use of MC, while in hospital. The hospital has a smoke-free policy for tobacco
products that prohibts smoking or vaping on hospital property. The hospital is currently
developing its MC policy and intends to treat MC like tobacco. The patient does not wish to
switch to a different route of administration, such as a synthetic cannabinoid or edibles given
his familiarity and prior success with inhaled MC. If applicable law has not clarified if MC
should be treated like tobacco in acute care settings, HCOs must decide whether or how they
might accommodate the patient’s use of MC both in the case at hand and more broadly through
HCO policy.
     Canada legalized MC in 2001 and implemented a major regulatory overhaul in 2013 by
enacting the “Marihuana for medical purposes regulations” (MMPR) (1, 2). The purpose of
the regulatory shift in 2013, which came into effect in 2014 via the MMPR, was to treat MC
similar to a narcotic (2, 3). Notably, the MMPR shifted the role of authorizing MC from Health
Canada to physicians (4). Since installing the MMPR, provincial bodies have begun to issue
physician practice standards to guide physician practice; however, such practice standards do
not address how healthcare organizations, like hospitals, should handle the use of MC (5, 6).
Therefore, unless provinces and territories pass MC legislation, HCOs must develop
institutional policy to address the use of MC.
     In August 2016, the MMPR was replaced by the “Access to cannabis for medical purposes
regulations” (ACMPR) (7). The main reason for the passage of ACMPR in 2016 was to
faciliate better access to MC by permitting individuals to register with Health Canada to grow
their own limited supply or to designate someone to produce it on their behalf (8). Additionally,
the regulations now also include fresh cannabis and cannabis oil, whereas the MMPR only
addressed dried cannabis (8). The new regulations also adopted the terminology of MC (9).
     As practicing healthcare ethicists in large academic and community acute care settings, we
have been consulted both for individual patient MC cases such as the illustrative one above as
well as on institutional policy development. Based on our experience to date, for Canadian
healthcare institutions that are developing a MC policy, we suggest that there are five broad
policy considerations that should be contemplated, which include the following: 1) clinical
management; 2) locus of administration; 3) route(s) of administration; 4) supply security; and
5) the therapeutic relationship. Additionally, in the context where applicable legislation is
evolving or provides incomplete direction, we have identified several associaed ethical issues
that warrant consideration; namely, harm reduction and key principles such as respect for
autonomy, non-malificence, reciprocity, justice and the harm principle (10). In the following
we will explore each of the five policy considerations and associated ethical issues in the
Canadian MC context.
               Ethical and policy implications concerning medical cannabis                     201
                                        DISCUSSION
The CPSO marihuana for medical purposes policy directs physicians when considering whether
they want to prescribe MC for a patient to prudently weigh whether MC is the most appropriate
treatment for their patient, and weigh the available evidence in support of dried cannabis against
other available treatment options (5). Additionally, because current evidence suggests that
children, adolescents and young adults have an increased risk of negative effects, physicians
are directed not to prescribe dried cannabis to patients under the age of 25 years unless all other
conventional therapeutic options have been attempted and have failed to alleviate the patient’s
symptoms (5). Physicians are also directed to advise patients about the material risks and
benefits of dried cannabis, such as operating a motor vehicle (5). The CPSO also recommends
that physicians require patients for whom they are prescribing MC to sign a written treatment
agreement (5). This type of written agreements are common for patients who are prescribed
opiods.
     In the acute care context, clinical management of MC may include either initiating or
authorizing continued use of MC and monitoring for side-effects. Authorization for continued
use is necessary since continuting therapy in the acute care context (typically) requires a
physician order. In our experience, it is rare that MC use is initiated in the acute care context;
rather, it is more common for physicians to be asked by patients to authorize the continued use
for MC prescribed previously in the community. In the event of the latter, the Most Responsible
Physician is charged with deciding whether they wish to authorize the continued use of MC
while the patient is in hospital.
     Additionally, clinical managment may involve overseeing a change in administration
modes. For example, if we reconsider our illustrative case, if the hospital has a policy
prohibiting smoking or vaping (either tobacco or MC, or both) within the hospital, a switch to
another mode of administration, such as ingesting MC, may be required.
202                               Sally Bean and Maxwell J Smith
Locus of administration
Institutional and community settings pose unique practice concerns. For example, allowing
smoking or vapourizing in a public hospital could contravene provincial smoke-free and
workplace health and safety legislation as well as institutional policy. In the community setting,
e.g., there may be applicable workplace legislation protecting home health workers; however,
there generally has to be greater flexibility since care is being provided in the patient’s home.
Routes of administration
There are four main options for of the route of administration for MC. Administration by
smoking offers the benefit of being rapidly absorbed because the inhaled route allows for real-
time dose titration (12). However, smoking MC produces second-hand smoke and commonly
a pungent odor. There is little evidence regarding the potential harms of second-hand smoke of
MC (13). Administration of MC by vapourizing mirrors the absorption and titration of smoking
MC, and the evidence of the potential harms of second-hand vapour of MC is more scant than
for smoking MC (12). Ingesting MC through edibles or tea produces a delayed onset (from 30-
120 minutes), which can make dose titration more challenging but does not produce second-
hand smoke or vapour (12). Finally, there are oral synthetic preparations (i.e., Cannabinoids)
that can be prescribed. Cannabinoids are pharmaceutically engineered, cannabis alternatives
and contain the active ingredient in marijuana, THC (14).
Security
Within the acute care setting, avoiding the abuse, misuse and diversion of MC supply through
appropriate storage is a key liability and policy concern. This is typically addressed by
establishing a policy or guideline for healthcare provider involvement with the administration
of MC as well as use of patient waiver or consent forms.
Although stigma around the use of MC may linger, it must be acknowledged that MC is a
permitted medical treatment and that individuals using it should not be stigmatized or
unreasonably disadvantaged by institutional policy. Integrating the ethical principles outlined
below may militate against these negative outcomes in HCO policy development. Additionally,
given the positive benefits of a healthy trust-based relationship, such as treatment adherence,
enduring relationships and perceived effectiveness of care, it is important that physicians and
patients maintain a healthy trust-based relationship that is not undermined by stigma or a
burdensome or unfair HCO policy (15).
               Ethical and policy implications concerning medical cannabis                    203
                                     ETHICAL ISSUES
With a limited evidence-base on negative effects of second hand smoke from MC, HCOs ought
to consider relevant ethical considerations and principles to help inform policy decision-making
for MC. We first consider potential contributions from the harm principle and the harm
reduction paradigm.
Harm principle
Anti-smoking interventions are (largely) justified by the harm principle, which affirms that
individuals are free to self-abuse unless harms accrue to others (16). The harm principle itself
generally posits that ‘. . . the only purpose for which power can be rightfully exercised over any
member of a civilized community, against his will, is to prevent harm to others’ (17). When
applied in the smoking context, this means that the negative effects of second-hand smoke
override the right of the smoker to smoke around others (16, 10) As such, HCOs may choose
to prohibit the use of MC on similar grounds. However, smoking is just one route of
administration for MC. Additionally, there is very little evidence regarding MC and potential
harm to others. One study, for instance, demonstrated that second hand smoke can produce
levels of THC in blood and urine and induce physiological effects (13). However, there is very
limited evidence and further research is required to make an evidence-based policy decision
stricly on these grounds.
Harm reduction
Harm reduction is to implement a measure, action or policy that has less harmful consequences
than a more harmful behaviour (16). The goal within a harm reduction paradigm is to introduce
a measure, action or policy that satisfies the need or behaviour associted with the harmful action
but that produces less harmful side effects or direct harm (16, 10). It is noteworthy that MC is
not associated with mortality, unlike opioids, which may be prescribed for similar conditions
(4). For example, a JAMA article noted a 25% reduction in opioid-related overdose fatalities
in states within the United States that enacted MC laws (4, 18). While it is unclear if this is a
correlative or causal relationship, the potential for harm reduction over opioids cannot be
overlooked.
     In addition to the insights generated from considering to the harm principle and harm
reduction paradigm, we can also draw upon complementary ethical principles of respect for
autonomy, non-maleficence, reciprocity and justice to further guide policy development in this
arena.
     The principle of respect for autonomy reflects the importance of patient self-determination.
HCOs that place significant restrictions on MC use may impair patients’ abilities to implement
a treatment regimen that best aligns with their preferred therapeutic goals. Additionally, overly
burdensome restrictions on legally available therapeutic options may undermine patient’s
abilities to implement potential harm reduction measures (relative to opioids).
204                               Sally Bean and Maxwell J Smith
     The principle of non-maleficence or doing no harm contributes the idea that if a patient has
received authorization to use MC then changing their route of administration (i.e., from inhaling
to ingesting) may unnecessarily harm the patient (i.e., produce less desirable health outcomes).
     The principle of reciprocity suggests that if we restrict or take away something away that
an individual is otherwise entitled (e.g., the use of MC in a form of administration as previously
authorized), we have an obligation to limit or mitigate the burden generated by that action.
Since there are other routes of administration of MC, some may argue that alternatives to
smoking or vaping, such as edibles or synthetic cannabinoids, meet this requirement. However,
that application is in tension with the principle of non-maleficence and autonomy if an
alternative route of administration is perceived to be less efficacious or if the patient does not
wish to switch routes of administration.
     Consideration of justice supports the idea that like cases should be treated alike and
dissimilar cases should be treated in a way that reflects relevant dissimilarities. Inconsistent
policy approaches across a patient population, or across HCOs, may foster injustice if a patient
using MC is treated different based on arbitrary reasons. Additionally, MC being held to more
stringent standards than other pain-relieving drugs with potential harmful effects broaches
notions of injustice if the different treatment cannot be justified by dissimilarities relative to
comparators.
                                    POLICY OPTIONS
For HCOs considering a policy approach to MC, we believe there are three broad policy options
for routes of administration:
handle MC smoke or vapour differently than tobacco smoke or vapour and enforce a policy
banning the latter but allowing the former.
     Option 3—to prohibit smoke or vapour producing routes of MC administration indoors
only—has the benefit of allowing patients that are able to ambulate individually or with
assistance to still continue their preferred route of administration without potentially harming
others. Similar to option 2, if in a jurisdiction with tobacco or smoke-free legislation, this also
may not be feasible or it may be practically difficult to enforce a different standard to MC
versus tobacco.
     In addition to the overarching route of administration questions, some common questions
warrant further consideration in HCO MC policy development given the brief discussion above:
                                       LIMITATIONS
Although many of the policy and ethical considerations are transferable to other contexts, this
paper has focused on the Canadian context which may differ from other jurisdicitons.
                                       CONCLUSION
In the backdrop of current MC discussions in Canada is the stated intention of Canadian Prime
Minister, Justin Trudeau, to introduce legislation in the Spring of 2017 to legalize recreational
use of cannabis (19). As a result, some may question the need for a HCO to create a MC policy
if such legislation is passed. While the contents of the draft legislation to be introduced are
hitherto unknown, it is safe to assume that there will be some use parameters for recreational
cannabis use that may potentially be differentiated from MC use, and that these parameters will
likely require careful consideration within HCOs.
     In this brief review we have highlighted a number of policy and ethical considerations for
HCOs that are now charged with developing MC policy. While additional research on MC
routes of administration and potential harm via MC second hand smoke or vapour is required
in order to make evidence-informed policy, the considerations and options discussed in this
206                                   Sally Bean and Maxwell J Smith
article provide a starting point for HCOs wishing to develop policy to address MC use until
relevant legislation is passed.
                                            REFERENCES
[1]    Minister of Justice. Marihuana for Medical Purposes Regulations, SOR/2013-119. Government of
       Canada Department of Justice 2015. URL: http://www.laws-lois.justice.gc.ca/PDF/SOR-2013-119.pdf.
[2]    Spithoff S. Emerson B. Spithoff A. Cannabis legalization: adhering to public health best practice. CMAJ
       2015; 187(16): 1211-6.
[3]    Canadian Medical Protective Association. Medical marijuana: considerations for Canadian doctors.
       Canadian Medical Protective Association 2016. URL: https://www.cmpa-acpm.ca/en/duties-and-
       responsibilities/-/asset_publisher/bFaUiyQG069N/content/medical-marijuana-new-regulations-new-
       college-guidance-for-canadian-doctors.
[4]    Lake S, Kerr T, Montaner J. Prescribing medical cannabis in Canada: Are we being too cautious? Can
       J Public Health 2015; 106(5): e328–30.
[5]    College of Physicians and Surgeons of Ontario. Marijuana for medical purposes – policy statement
       #1-15. Toronto, ON: College of Physicians and Surgeons of Ontario, 2015.
[6]    College of Physicians and Surgeons of British Columbia. Marijuana for medical purposes - professional
       standards and guidelines. Vancouver,       BC: College of Physicians and Surgeons of British
       Columbia, 2015.
[7]    Government of Canada. Access to cannabis for medical purposes regulation, (SOR/2016-230). Canada
       Gazette 2016. URL: http://gazette.gc.ca/rp-pr/p2/2016/2016-08-24/html/sor-dors230-eng.php.
[8]    Laviolette C. Wakulowsky L. Guidance for health care organizations on the new cannabis regulations.
       Borden Ladner Gervais 2016. URL: http://www.blg.com/en/NewsAndPublications/Publication_4636
[9]    Vallerihani J. New pot rules a welcome step on the road to legalization. Globe and Mail 2016. URL:
       http://www.theglobeandmail.com/opinion/new-pot-rules-a-welcome-ste p-on-the-road-to-legalization/
       article31381947/.
[10]   Bean S. Smith M. A vaping matter: e-cigarette use in health care organizations. Hastings Cent Rep 2015:
       45(6): 11-12.
[11]   Hall W. U.S. policy responses to calls for the medical use of cannabis. Yale J Biol Med 2015; 88(3):
       257-64.
[12]   Wilkinson ST, Yarnell S, Radhakrishnan R. Ball SA, D’Souza DC. Marijuana legalization: impact on
       physicians and public health. Annu Rev Med 2016; 67: 453-66.
[13]   Herrmann ES, Cone EJ, Mitchell JM, Bigelow GE, LoDico C, Flegel R, et al. Non-smoker exposure to
       secondhand cannabis smoke II: Effect of room ventilation on the physiological, subjective, and
       behavioral/cognitive effects. Drug Alcohol Depend 2015; 151: 194-202.
[14]   Canadian Medical Association. Medical marijuana. Canadian Medical Association 2011. URL:
       http://policybase.cma.ca/dbtw-wpd/Policypdf/PD11-02.pdf.
[15]   Shea K, Effken J. Enhancing patients’ trust in the virtual home healthcare nurse. Comput Inform Nurs
       2008; 26: 135-141.
[16]   Holland S. Public health ethics, 2nd ed. Cambridge, UK: Polity Press, 2007.
[17]   Mill JS. On liberty and other essays. New York: Kaplan Publishing, 2009.
[18]   Bachhuber MA, Saloner B, Cunningham CO, Barry CL. Medical cannabis laws and opioid analgesic
       overdose mortality in the United States, 1999–2010. JAMA Intern Med 2014; 174(10): 1668–73.
[19]   Federal marijuana legislation to be introduced in spring 2017, Philpott says. Canadian Broadcasting
       Corporation News 2016. URL: http://www.cbc.ca/news/politics/philpott-un-marijuana-legislation-
       legalize-1.3544554.
In: Cannabis: Medical Aspects                                            ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                   © 2017 Nova Science Publishers, Inc.
Chapter 17
    Cannabis is a drug that has been around since ancient times and continues to be one of the
    most popular drugs used worldwide today. Even though cannabis is looked at as a low risk
    drug with medicinal benefits, there are actually many concerning adverse effects related to
    marijuana use. Research shows that there are various psychiatric, neurological, respiratory,
    endocrine, and gastrointestinal complications that can be caused by cannabis use. Medical
    providers need to be aware of these various adverse effects, especially when caring for the
    adolescent population.
                                         INTRODUCTION
Due to the increase in medicinal use and legalization of recreational use in some US states and
many countries around the world, marijuana is a drug that has made its way back into the
spotlight. Marijuana has been around since ancient times and has been mainly used to achieve
euphoria. The medicinal purposes of marijuana are currently being studied and touted by some,
but due to the complex legal atmosphere surrounding the therapy it has not been widely
prescribed. Recreational use of the drug continues at very high rates. The supply of marijuana
is more abundant than ever before, making it much easier for the adolescent population to
obtain.
    Adolescents are not only experimenting with marijuana, but some are using it in various
forms on a daily basis. In recent surveys, teens admit that marijuana is easy to obtain (1). In
2014, 21% of high school seniors reported having tried marijuana and almost 6% of high school
seniors reported daily marijuana use (1). One of the most concerning survey findings was that
 Correspondence: Amy L Burnett, MSN, APRN, Adolescent Medicine, University of Kentucky College of Medicine,
    740 S Limestone, Lexington, KY 40536, United States. E-mail: amy.burnett1@uky.edu.
208                                        Amy L Burnett
the perception of risk associated with the use of marijuana in the adolescent population has
consistently declined over the past few years (1). Adolescents not only feel that there is no risk
in marijuana use, but can often endorse benefits from marijuana use (2, 3). The purpose of this
article is to explore the various adverse effects associated with cannabis use in the adolescent
population.
                                    ADVERSE EFFECTS
Most people are aware of the acute adverse effects caused by marijuana, or cannabis, ingestion.
The physical symptoms of conjunctival injection, increased appetite, dry mouth, and
tachycardia are seen in most individuals within two hours of use (1). These physical symptoms
accompany the psychological symptoms of euphoria, which can present differently across
individuals. Some are very relaxed and sedate, while others develop more of an anxious
paranoia. These acute symptoms typically fully resolve within a few hours of cannabis use.
     Chronic cannabis use can lead to more significant, and sometimes irreversible, adverse
effects. The most studied adverse effects of chronic cannabis use are the effects that the drug
has on the brain.
     Cannabis is known to cause short-term memory impairment, decrease concentration and
attention span, and altered problem solving capabilities (4). Most of those that use marijuana
are under the assumption that the drug decreases anxiety, when in reality 20% to 30% of users
report an increase in generalized anxiety and panic attacks (5). There is conflicting research as
to the effects that cannabis use has on depression and if there is a causal relationship, but it is
thought that marijuana may worsen existing depression (5). Cannabis use has been associated
with increased rates of psychosis in those with a schizophrenia diagnosis and in those that are
genetically predisposed to schizophrenia (6). Due to an immature prefrontal cortex in the
adolescent, marijuana use is particularly concerning during the teenage years. It is uncertain
what impact this drug has on the developing brain, but research has shown that chronic cannabis
use has been associated with lower high school completion rates and lower college degree
attainment (7).
     It goes without saying that there are adverse respiratory affects associated with cannabis
use, seeing as the most common form of use is inhalation. Cannabis use has been linked to
increased irritation, swelling, and secretions of the airways, leading to more frequent respiratory
infections, cough, asthma, and chronic bronchitis (8). Chronic cannabis use has been associated
with the development of chronic obstructive pulmonary disease later in life. It has been difficult
to conclusively link long-term marijuana smoking to lung cancer, but it is thought that there
could be a causal relationship (9).
     There have been many noted effects on the endocrine system of the body and chronic
cannabis use. Cannabis causes an imbalance in the sex hormone production in both males and
females, leading to a host of reproductive issues (10). Cannabis use lowers testosterone levels
in males, which can result in decreased libido and sperm count and gynecomastia in the male
(11). Chronic use has been shown to increase prolactin levels in females, resulting in
galactorrhea in some chronic users (10). Cannabis use has also been found to inhibit growth
hormone secretion, thyroid function, and glucose levels (10).
                                Adverse effects of cannabis use                                    209
    Another adverse effect of chronic cannabis use that has come to light in recent years is
cannabinoid hyperemesis syndrome (CHS). This syndrome is characterized by cyclic nausea,
vomiting, and epigastric pain (12). These side effects resolve with taking hot showers and the
cessation of marijuana use, but do not typically respond to anti-emetics (13). With chronic
cannabis use, disequilibrium within the hypothalamus occurs, causing issues with
thermoregulation. It is thought that being in hot water helps to correct this imbalance (13). With
CHS, patients will typically have a decrease in symptoms when they stop cannabis use and
should fully resolve after about a week of complete cessation (14). The symptoms of CHS can
recur if the individual starts using cannabis again in the future (14).
    Not only does cannabis use cause unintended adverse effects, the cessation of use can also
create a host of unwanted symptoms. Cannabis withdrawal has been well studied and many
physical and psychological withdrawal symptoms have been identified. Individuals will
commonly present with the psychological symptoms of anger, aggression, anxiety, restlessness,
insomnia, irritability, and depressed mood (15). Physical symptoms of cannabis withdrawal of
decreased appetite, weight loss, chills, abdominal pain, shakiness, and sweating have also been
noted with cannabis withdrawal (15). It is thought that around 67% of adolescents who are
cannabis dependent will develop withdrawal symptoms with cessation of the drug (16). The
onset of these symptoms can occurs within the first 1-3 days and can last up to 14 days after all
cannabis use has stopped (17).
                                         CONCLUSION
Cannabis is a drug that continues to grow in popularity and use. It is now being marketed as a
treatment for many illnesses and is being legalized for recreational use in some states and
countries around the globe. While the medicinal benefits sound appealing, medical providers
must be aware of the various adverse effects caused by cannabis use, especially in the
adolescent population. With more legal marijuana in circulation, adolescents will have much
more exposure to, and use of, this drug. Those providing care to this patient demographic need
to be aware of all potential dangers of cannabis use in order to properly care for the adolescent
population.
                                         REFERENCES
[1]   High school and youth trends. Drugabuse.gov. 2016. URL: https://www.drugabuse.gov/
      publications/drugfacts/high-school-youth-trends.
[2]   Roditis ML, Halpern-Felsher B. Adolescents’ perceptions of risks and benefits of conventional
      cigarettes, e-cigarettes, and marijuana: A qualitative analysis. J Adolesc Health 2015;57:179-85.
[3]   Krader CG. Why AAP opposes marijuana use. Contemp Pediatr 2016 Febr 01.
[4]   Harvard Medical School. Medical marijuana and the mind. Harv Ment Health Lett 2010;26:1-3.
[5]   Volkow ND, Baler RD, Compton WM, Weiss SR. Adverse health effects of marijuana use. N Eng J
      Med 2014;370:2219-27.
[6]   American Academy of Pediatrics. The impact of marijuana policies on youth: Clinical, research, and
      legal update. Pediatrics 2015;135:584-7.
[7]   Tashkin DP, Baldwin GC, Sarafian T, Dubinett S, Roth MD. Respiratory and immunologic
      consequences of marijuana smoking. J Clin Pharmacol 2002;42:71S-81S.
210                                           Amy L Burnett
[8]    Joshi M, Joshi A, Bartter T. Marijuana and lung diseases. Curr Opin Pulm Med 2014;20:173-9.
[9]    Brown TT, Dobs AS. Endocrine effects of marijuana. J Clin Pharmacol 2002;42:90S-6.
[10]   Kolodny RC, Masters WH, Kolodner RM, Toro G. Depression of plasma testosterone levels after
       chronic intensive marihuana use. N Engl J Med 1974;290(16):872.
[11]   Beech RA, Sterrett DR, Babaiuk J, Fung H. Cannabinoid hyperemesis syndrome: A case report and
       literature review. J Oral Maxillofac Surg 2015;73:1907-19.
[12]   Heise L. Cannabinoid hyperemesis syndrome. Adv Emerg Nurs J 2015;37:95-101.
[13]   Wilson O, Lutton S, Doherty K. Diagnosing and treating cannabinoid hyperemesis. Emerg Nurs
       2015;23:22-5.
[14]   Budney AJ, Hughes JR, Moore BA, Vandrey R. Review of the validity and significance of cannabis
       withdrawal syndrome. Psychiatry 2004;161:1967-77.
[15]   Kouri EM, Pope HG, Lukas SE. Changes in aggressive behavior during withdrawal from long-term
       marijuana use. Psychopharmacol 1999;143:302-8.
[16]   Haughey HM, Marshall E, Schacht JP, Louis A, Hutchinson KE. Marijuana withdrawal and craving:
       influence of the cannabinoid receptor I (CNRI) and fatty acid amide hydrolase (FAAH) gene. Addict
       2008;103:1678-86.
In: Cannabis: Medical Aspects                                                ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                       © 2017 Nova Science Publishers, Inc.
Chapter 18
            Despite all the efforts to prevent and remediate adolescent drug use, the use of
       marijuana among adolescents remains both high and stable. Of equal concern, more and
       more adolescents think that occasionally smoking marijuana is not harmful. In this chapter
       we aim to discuss the role of schools in combating adolescent use of marijuana and possible
       interventions through the Multi-Tier Systems of Support (MTSS).
                                             INTRODUCTION
Marijuana is the most popularly used illicit drug among adolescents (1, 2). According to the
Monitoring the Future study (national survey of 8th, 10th, and 12th grade students), about 24%
of adolescents reported that they used marijuana in the past year (1). Nationwide, 14% of
students reported they used marijuana one or more times within 30 days before the survey (1).
The use of marijuana among adolescents remains stable, while the use of alcohol, cigarettes,
and prescription pain relievers is declining (1). Of equal concern is that more and more
adolescents think that occasionally smoking marijuana is not harmful (1).
    During adolescence, young individuals go through many physical, cognitive,
psychological, and social changes. As discussed below, the use of marijuana puts the
transforming body and mind at risk. Adolescent use of marijuana has been linked to a variety
of short- and long-term problems.
*
    Corresponding author: Alissa Briggs, PhD, NCSP, Assistant Professor and Licensed Psychologist, UK Healthcare,
       Department of Pediatrics, Division of Adolescent Medicine and Young Parents Program, KY Clinic, Room
       L443, Lexington, KY 40536-0284, United States. E-mail: alissa.briggs@uky.edu.
212                                Venus Wong and Alissa Briggs
Research found that persistent use of marijuana during adolescence impairs cognitive
functioning (3-5). A study found that persistent adolescent-onset marijuana users have
significantly weaker working memory, processing speed, perceptual reasoning, and verbal
comprehension, when compared to non-marijuana users, after controlling for years of education
and childhood cognitive ability (4). Such a global cognitive impairment is more prevalent
among adolescent-onset marijuana users than adult-onset users (4).
    Not surprisingly, use of marijuana is associated with low school performance, which may
be a result of declining cognitive ability. Research found that use of marijuana predicted years
of education (6), high school dropout (7), perceived academic failure (8), low grades (9), and
academic achievement (10). However, the relationship between the use of marijuana and low
school performance appears to be complex (9). Some studies treated low school performance
as a predictor of the use of marijuana. A number of studies found that negative educational
outcomes precede marijuana use (11-13). Some other evidence supports the idea that a third
variable accounts for the use of marijuana and negative education outcomes (9, 14).
associated behavioral and mental health issues usually partake in partial hospitalization or
inpatient treatment (26). That is, many direct interventions for adolescents with problems
associated with marijuana use are handled by non-school professionals from outside agencies.
At times, students who exhibit more severe marijuana use and associated behavioral and mental
health issues are placed in a more structured environment (e.g., hospital) and do not attend
regular schools until their problems are relieved.
     Even though typical schools may not possess sufficient resources to support students with
current and severe addiction problems, they have unique advantages in two areas across the
spectrum of care. First, schools play an important role in the early identification of marijuana
use (28). Teachers are in a good position to observe marijuana-use related symptoms, such as
changes in mood, academic performance, behaviors, and physical appearance (29). Early
detection benefits students because it usually leads to early interventions that prevent further
damage as a result of the drug use (30, 31). Second, since the majority of adolescents attend
school on a regular basis, schools have the advantage of delivering prevention education for
the general student population, supplementary services, and relapse prevention services.
Adolescent drug abuse prevention and early intervention programs can be categorized into three
levels: universal, selective, and indicated programs to (32). Universal programs target the
general adolescent population, with the aim of preventing or delaying the use of drugs. This
type of program addresses common risk and protective factors (e.g., peer pressure, learning
difficulties) (32). Selective programs are more restricted and target high-risk adolescent groups
(e.g., students who display a variety of risk-taking behavior, students whose parents abuse
drugs, students who are diagnosed with anxiety or depression). The aim of selective programs
is to provide students with more intensive, specific support to make responsible, healthy
decisions (32). Indicated programs are designed for adolescents who have used drugs. The aims
of indicated programs are to motive adolescents to reduce the use of drugs and diminish the
negative outcomes associated with drug use (32). Indicated programs should cover all forms of
drug abuse (e.g., use of illegal drugs, inappropriate use of legally obtained drugs, underage
alcohol consumption), even though these programs should also be tailored to address specific
drug use problems and risk factors (1, 33, 34).
214                               Venus Wong and Alissa Briggs
    Schools also use a a three-tiered model of service delivery. Within schools, a three-tiered
model of service delivery is referred to as Multi-Tier System of Supports (MTSS). Multi-Tier
System of Supports is used as a vehicle to coordinate prevention and intervention efforts for a
wide variety of concerns (e.g., academic, behavioral, and social-emotional) (35) and can target
drug use. Moreover, MTSS is a data-based decision making model (36) that involves parents.
Within MTSS, universal screening and ongoing data collection are implemented periodically
in order to provide information about intra-and inter-student needs and progress (37). Parents
should be involved in the planning and intervention process (38). Furthermore, involving
parents in interventions can ensure continuing quality support outside the school and prompt
generalization of skills. The nature of the MTSS echoes the needs for parental involvement and
on-going monitoring for students who use marijuana (see (39, 40)).
Tier 1
Description. Tier-one interventions target the whole student population, with the aim of
preventing or delaying the use of marijuana and other drugs. The target of this group should be
drug-users and non-drug users (26). Universal programs are generally less expensive per
student, shorter in length, less intensive, include all youths in schools, and do not require trained
professionals for implementation (41). Yet, because of the general nature of the programs, they
usually do not meet individual needs. Also, the intensity of these programs is not sufficient to
eliminate the negative effects of risk factors.
     Other common effective practices. Teachers also need to keep a close eye on students
when they are in school. A national report found that around 6% of students admitted using
marijuana on school property at least one time within the 30 days before the survey (2). Close
monitoring of student behaviors (e.g., during transition, recess) is needed in order to ensure
drug-free schools.
     Ongoing assessment. As part of MTSS, schools should be regularly (usually monthly)
reviewing data in order to identify students who are at-risk. As aforementioned, marijuana use
is associated with a variety of negative educational outcomes, and thus promptly identifying
students who are at-risk educationally may help interrupt the likely bi-directional relationship
between marijuana use and academic failure. Moreover, some schools regularly screen for signs
of depression in an effort to prevent suicide, and such screening could also help identify
students at-risk for marijuana use.
     Parent involvement. Parents are an important factor in the prevention of marijuana use
(39, 42). One of the main aims at tier one is to increase parents’ awareness of drug use issues
in their communities and empower them with the knowledge necessary for them to identify the
use of drugs. All parents should be given information with regard to drug prevention and the
signs of marijuana use and addiction. Parents should also be clearly informed about the steps
they need to take if they suspect their child of using drugs. Since research has found that parent-
child relationships (e.g., closeness, warmth) and parental monitoring (e.g., parental knowledge
of the child’s whereabouts, friendships, activities, and use of money) are two important
predictors of adolescent drug use (39), teachers could support parents in incorporating effective
parenting practices when communicating with parents through newsletters, school-to-home
notebooks, monthly phone calls, and, if appropriate, parent-teacher conferences (42). Schools
could also facilitate positive parental involvement through (1) shared governance on advisory
committees; (2) active, two-way communication; (3) encouraging parents to plan and attend
school events; (4) assisting teaching in the classroom; and (5) providing parent education
activities (43).
Tier 2
     Ongoing student assessment. Data monitored at tier one should be monitored on a more
regular basis for students at tier 2 (e.g., bi-monthly). In addition, regular assessment of at-risk
students could be implemented by qualified school professionals (e.g., school nurse, school
psychologist) using tools such as the Universal Screening for Substance Use, Brief
Intervention, and/or Referral to Treatment (SBIRT; (44)) and the CRAFFT screening (45).
     Parent involvement. At this stage, home-school collaboration is critical so that parents
and school personel can work together to support high-risk adolescents. The first step in the
process is for school personnel provide parents with data, inform them of their concerns, and
develop a plan of action. At this stage, parents need to be aware of and even consent to
additional supports put in place for their children and should be informed their progress. They
also must have the opportunity to help develop a support plan and reinforce interventions
provided at home.
Tier 3
Description. Tier-three interventions target adolescents who have already begun using, or are
experimenting with marijuana. Services provided at tier three may be provided through
alternative schools or recovery schools. The main goals of this tier are to motivate adolescents
to reduce their use of marijuana and diminish the negative outcomes associated with marijuana
use (32). Services at this level should target students who are also receiving regular outpatient
or intensive outpatient services from outside agencies with regard to their marijuana use and
any other associated problems. Some students at tier three may be students who are released
from partial hospitalization and inpatient services. For this group of students, the services in
school can also aim to prevent relapse.
     Compared to tier-one and tier-two programs, programs at tiers three are more specific and
intensive. For instance, the Reconnecting Youth program is a daily curriculum that consists of
75 session and targets students who show low academic performance, have potential for
dropping out of high school, and exhibit other problem behaviors patterns such as substance
abuse. This program should only be conducted by a trained professionals (46).
     Other effective practices. Since students at tier three may have recently been released
from more restricted inpatient drug treatment programs, this stage can also be considered a
transition. Before the student is transferred back to the school, school professionals, parents,
and staff from the inpatient unit should develop an individualized transition plan. The
continuity of services is the key to student success (47). Schools need to ensure good
communication with both parents and outpatient units about the goals of the transition and the
steps needed to achieve these goals. Quality transition goals need to be specific, measurable,
achievable, realistic/relevant, and timed (48). The transition plan also needs to specify clearly
the roles of each party and the services needed by the family.
     The MTSS system is designed in such a way as to provide the majority of the
aforementioned components early on (e.g., encouraging family involvement; increasing
prosocial leisure habits). However, compared to students without drug use, students at tier three
need extra intense, targeted, and structured opportunities to learn prosocial behaviors. The
aforementioned components can be a guideline for schools to develop tier- three relapse
prevention support.
                             Cannabis and the role of our schools                                   217
     Ongoing student assessment. Since students at this stage have already started
experimenting or using drugs regularly, they usually partake in outside services. Despite the
relatively high need for support, students at tier three usually still spend a significant amount
of time in school. Frequent communication between the school and outside professionals will
facilitate data-based decision making, provided that the parents and adolescents concsent to
such collaboration. Assessment that takes place at tier two should continue to take place, but
more frequently (e.g., weekly). In addition, assessment at tier three may also be used for relapse
monitoring. Some simple progress monitoring tools, such as the Routine Outcome-Monitoring
(ROM) questionnaire, can be used regularly after drug abuse treatment (49).
     Parent involvement. At this stage, students are usually supported by outside agencies. For
students who are transferred back to school from inpatient programs, the school, outpatient unit,
and parents should collaborate on the transition planning and interventions. It is important to
note that parents at this stage may often seek advice, comfort, or encouragement from school
professionals (50). Teachers and school-based mental health professionals need to be prepared
to provide timely informational and emotional support for parents.
                                         CONCLUSION
Adolescent use of marijuana is a pervasive problem in society. Schools can play a critical in
addressing this problem. Much previous work has developed effective universal, selected, and
indicated marijuana and other drug prevention programs. It is important to note that despite the
established effectiveness, these evidence-based programs suffer from limitations. In face of the
idiosyncratic challenges experienced by different communities, it is important for schools to
select the right drug prevention programs and develop their own anti-drug strategies based on
existing research findings and their particular situations. This paper clearly delineates the utility
of the MTSS model in school as a service delivery framework for students with or without
marijuana use problems, and how MTSS connects with outside service delivery systems.
Schools are encouraged to use the MTSS model to guide their drug prevention implementation,
provide universal screening, make data-based decisions, and involve parents in the prevention
process.
                                         REFERENCES
[1]   Johnston LD, O’Malley PM, Miech RA, Bachman JG, Schulenberg JE. Monitoring            the    Future
       national survey results on drug use, 1975-2015: Overview, key findings on adolescent drug use. Ann
       Arbor, MI: Institute Social Research, 2016.
[2]   Centers for Disease Control and Prevention. Alcohol- or other drug-use prevention, 2012 URL:http://
      www.cdc.gov/healthyyouth/shpps/2012/factsheets/pdf/FS_AlcoholOrOtherDrugUsePrevention_
      SHPPS2012.pdf.
[3]   Dougherty DM, Mathias CW, Dawes MA, Furr RM, Charles NE, Liguori A, et al. Impulsivity, attention,
      memory, and decision-making among adolescent marijuana users. Psychopharmacology (Berl) 2013;
      226(2): 307-19.
218                                    Venus Wong and Alissa Briggs
[4]    Meier MH, Caspi A, Ambler A, Harrington H, Houts R, Keefe RS, et al. Persistent cannabis users show
       neuropsychological decline from childhood to midlife. Proc Natl Acad Sci USA 2012; 109(40): E2657-
       64.
[5]    Zalesky A, Solowij N, Yucel M, Lubman DI, Takagi M, Harding IH, et al. Effect of long-term cannabis
       use on axonal fibre connectivity. Brain 2012; 135(Pt 7): 2245-55.
[6]    Chatterji P. Illicit drug use and educational attainment. Health Econ 2006; 15(5): 489-511.
[7]    Bray JW, Zarkin GA, Ringwalt C, Qi J. The relationship between marijuana initiation and dropping out
       of high school. Health Econ 2000; 9(1): 9-18.
[8]    Bergen HA, Martin G, Roeger L, Allison S. Perceived academic performance and alcohol, tobacco and
       marijuana use: longitudinal        relationships in young community adolescents. Addict Behav 2005;
       30(8): 1563-73.
[9]    McCaffrey DF, Pacula RL, Han B, Ellickson P. Marijuana use and high school dropout: the influence
       of unobservables. Health Econ 2010; 19(11): 1281-99.
[10]   Brook JS, Stimmel MA, Zhang C, Brook DW. The association between earlier marijuana use and
       subsequent academic achievement and health problems: a longitudinal study. Am J Addict 2008; 17(2):
       155-60.
[11]   Horwood LJ, Fergusson DM, Coffey C, Patton GC, Tait R, Smart D, et al. Cannabis and depression: an
       integrative data analysis of four Australasian cohorts. Drug Alcohol Depend 2012; 126(3): 369-78.
[12]   Ledoux S, Miller P, Choquet M, Plant M. Family structure, parent-child relationships, and alcohol and
       other drug use among teenagers in France and the United Kingdom. Alcohol Alcohol 2002; 37(1): 52-
       60.
[13]   Newcomb MD, Bentler PM. IMpact of adoelscent drug use and social support on problems of young
       adults: A longitudinal study. J Abnorm Psychol 1988; 97(1): 64-75.
[14]   Barnes GE, Barnes MD, Patton D. Prevalence and predictors of “heavy” marijuana use in a Canadian
       youth sample. Subst Use Misuse 2005; 40(12): 1849-63.
[15]   Kingree JB, Betz H. Risky sexual behavior in relation to marijuana and alcohol use among African-
       American, male adolescent detainees and their female partners. Drug Alcohol Depend 2003; 72(2):
       197-203.
[16]   Staton M, Leukefeld C, Logan TK, Zimmerman R, Lynam D, Milich R, et al. Risky sex behavior and
       substance use among young adults. Health Soc Work 1999; 24(2): 147-54.
[17]   Asbridge M, Hayden JA, Cartwright JL. Acute cannabis consumption and motor vehicle collision risk:
       systematic review of observational studies and meta-analysis. BMJ 2012; 344: e536.
[18]   Johnson V, White HR. An investigation of factors related to intoxicated driving behaviors among
       youth. J Stud Alcohol 1989; 50(4): 320-30.
[19]   Dawkins MP. Drug use and violent crime among adolescents. Adolescence 1997; 32(126): 395-405.
[20]   Green KM, Doherty EE, Stuart EA, Ensminger ME. Does heavy adolescent marijuana use lead to
       criminal involvement in adulthood? Evidence from a multiwave longitudinal study of urban African
       Americans. Drug Alcohol Depend 2010; 112(1-2): 117-25.
[21]   Sawyer MG, Arney FM, Baghurst PA, Clark JJ, Graetz BW, Kosky RJ, et al. The mental health of young
       people in Australia: key findings from the child and adolescent component of the national survey of
       mental health and well-being. Aust NZ J Psychiatry 2001; 35(6): 806-14.
[22]   Medina KL, Nagel BJ, Park A, McQueeny T, Tapert SF. Depressive symptoms in adolescents:
       associations with white matter volume and marijuana use. J Child Psychol Psychiatry 2007; 48(6): 592-
       600.
[23]   Musty RE, Kaback L. Relationships between motivation and depression in chronic marijuana users.
       Life Sci 1995; 56(23-24): 2151-8.
[24]   Comeau N, Stewart SH, Loba P. The relations of trait anxiety, anxiety sensitivity, and sensation seeking
       to adolescents’ motivations for alcohol, cigarette, and marijuana use. Addict Behav 2001; 26(6): 803-
       25.
                               Cannabis and the role of our schools                                         219
[25]   Arseneault L, Cannon M, Poulton R, Murray R, Caspi A, Moffitt TE. Cannabis use in adolescence and
       risk for adult psychosis: longitudinal prospective study. BMJ 2002; 325(7374): 1212-3.
[26]   National Institute on Drug Abuse. Principles of adolescent substance use disorder treatment: A research-
       based guide, 2014. URL: https://www.ncjrs.gov/App/Publications/ abstract.aspx?ID= 267089.
[27]   Pelosi N. Reducing risks of mental disorders. Am Psychol 1996; 51(11): 1128-9.
[28]   Griswold KS, Aronoff H, Kernan JB, Kahn LS. Adolescent substance use and abuse: recognition and
       management. Am Fam Physician 2008; 77(3): 331-6.
[29]   National Institute on Drug Abuse. Marijuana: Facts parents need to know, revised, 2011. URL:
       https://www.drugabuse.gov/sites/default/files/parents_marijuana_brochure.pdf.
[30]   Ali S, Mouton CP, Jabeen S, Ofoemezie EK, Bailey RK, Shahid M, et al. Early detection of illicit drug
       use in teenagers. Innov Clin Neurosci 2011; 8(12): 24-8.
[31]   Werner MJ, Joffe A, Graham AV. Screening, early identification, and office-based intervention with
       children and youth living in substance-abusing families. Pediatrics 1999; 103(5 Pt 2): 1099-112.
[32]   National Institute on Drug Abuse. Preventing drug abuse: The best strategy, 2014. URL:
       https://www.drugabuse.gov/publications/drugs-brains-behavior-science-addiction/preventing-drug-
       abuse-best-strategy.
[33]   Hawkins JD, Catalano RF, Arthur MW. Promoting science-based prevention in communities. Addict
       Behav 2002; 27(6): 951-76.
[34]   Oetting ER, Edwards RW, Kelly K, Beauvais F. Risk and protective factors for drug use among rural
       American youth. NIDA Res Monogr 1997; 168: 90-130.
[35]   New York State Department of Education. Minimum requirements of a response to intervention
       program (RtI), 2010. URL: http://www.p12.nysed.gov/specialed/RTI/ guidance/ instruction.htm.
[36]   Proctor SL, Meyers J. Best practices in primary prevention in diverse schools and communities. In:
       Thomas A, Harrison P, eds. Best practices in school psychology: Foundations, 6 ed. Bethesda, MD:
       National Association of School Psychologists, 2014: 33-47.
[37]   RTI Action Network. What is RTI? URL: http://www.rtinetwork.org/learn/ what/whatisrti.
[38]   Mellard DF, Stern A, Woods K. RTI school-based practices and evidence-based models. Focus Except
       Child 2011; 43: 1-15.
[39]   Steinberg L, Fletcher A, Darling N. Parental monitoring and peer influences on adolescent substance
       use. Pediatrics 1994; 93(6 Pt     2): 1060-4.
[40]   Moberg DP, Finch AJ. Recovery high schools: A descriptive study of school programs and students.
       J Groups Addict Recover 2008; 2: 128-61.
[41]   Kumpfer KL. Identification of drug abuse prevention programs. US Department of Health
       and Human Services. URL: http://archives.drugabuse.gov/about/organization/despr/hsr/da-pre/
       KumpferLitReviewPartB.html.
[42]   Graham-Clay S. Communicating with parents: Strategies for teachers. Sch Commun J 2005; 15(1): 117.
[43]   Lyons P, Robbins A, Smith A. Involving Parents: A handbook for participating in schools. Santa
       Monica, CA: System Development Corporation 1982.
[44]   Madras BK, Compton WM, Avula D, Stegbauer T, Stein JB, Clark HW. Screening, brief interventions,
       referral to treatment (SBIRT) for illicit drug and alcohol use at multiple healthcare sites: comparison at
       intake and 6 months later. Drug Alcohol Depend 2009; 99(1-3): 280-95.
[45]   Kulig JW, American Academy of Pediatrics Committee on Substance Abuse, Tobacco, Alcohol, and
       Other Drugs. The role of the pediatrician in prevention, identification, and management of substance
       abuse. Pediatrics 2005; 115(3): 816-21.
[46]   Eggert LL. Reconnecting youth: An indicated prevention program. Washington, DC: National
       Conference on Drug Abuse Research, 1996.
[47]   Dickard N, Downs T, Cavanaugh D. Recovery/relapse prevention in educational settings for youth with
       substance use and co-occuring mental health disorders. Washington, DC: Department Education, 2011.
220                                  Venus Wong and Alissa Briggs
[48]   Bovend'Eerdt TJ, Botell RE, Wade DT. Writing SMART rehabilitation goals and achieving goal
       attainment scaling: a practical guide. Clin Rehabil 2009; 23(4): 352-61.
[49]   Lennox RD, Sternquist MA, Paredes A. A simplified method for routine outcome monitoring after drug
       abuse treatment. Subst Abuse 2013; 7: 155-69.
[50]   Hamilton NL, Brantley LB, Tims FM, Angelovich N, McDougall B. Cannabis Youth Treatment Series,
       2001; 3. URL: http://adaiclearinghouse.org/downloads/Family-Support-Network-for-Adolescent-
       Cannabis-Users-CYT-Series-Vol-3-340.pdf.
In: Cannabis: Medical Aspects                                              ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                     © 2017 Nova Science Publishers, Inc.
Chapter 19
       Cannabis is not a new drug. In fact, the cannabis or hemp plant grows in most parts of the
       world and has a long history of both medical, as well as non-medical use. Little is
       documented on the use of cannabis in the first half of the century, however, despite legal
       sanctions consumption of cannabis increased notably in the 1960s and 1970s and usage
       seems not to have abated since that time. Its resurgence as a medicinal drug of interest
       came out of the AIDS pandemic and in 1999 two patients obtained permission by the
       Federal Government to smoke marijuana. Subsequently in 2000 the Courts ruled that
       Canadians had a right to use cannabis as a medicine, and in 2001 the Liberal government
       enacted the Medical Marihuana Access Regulations (MMAR), allowing cannabis for
       therapeutic purposes.
                                            INTRODUCTION
In discussing the potential medical uses of cannabis, it is important to remember that cannabis
is not a new drug. In fact, the cannabis or hemp plant grows in most parts of the world and has
a long history of both medical, as well as non-medical use (1-2). However, by the mid 19th
Century stronger and more reliable synthetic drugs, such as opioids and other clinically tested
and approved narcotics, began to form the mainstream pharmacological interventions of choice
in medicine. Scientific interest in cannabis as a medicine waned at the start of the 20th Century,
and among the reasons for this loss of favor were that the plant material was too variable in
    Correspondence: Mr Blair Henry, Ethicist, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto,
       ON Canada. E-mail: Blair.Henry@sunnybrook.ca.
222                   Blair Henry, Rachel McDonald, Stephanie Chan et al.
composition (strength), its shelf-life was too short and unpredictable, uncontrolled sources
meant reduced purity, as well as the changing legal climate (3-4).
     The legal framework for drug control in Canada was created in the early 1990s. By 1908,
all medicines, inclusive of tobacco and alcohol, were on the way to regulation and it would be
in the same year that the Opium Act created the first drug prohibition (5). Cannabis was
officially added to the Canadian drug prohibition law in 1923, making its production,
possession, and consumption illegal activities (6). Little is documented on the use of cannabis
in the first half of the century, however, despite legal sanctions consumption of cannabis
increased notably in the 1960 and 1970, and usage seems not to have abated since that time (5).
Its resurgence as a medicinal drug of interest came out of the AIDS pandemic. In 1999 two
patients obtained permission by the Federal Government to smoke marijuana and in 2000 the
Courts ruled that Canadians had a right to use cannabis as a medicine (7).
     In 2001, the Liberal government enacted the Medical Marihuana Access Regulations
(MMAR), allowing cannabis for therapeutic purposes (8). Under this framework, physicians
were allowed to authorize cannabis use to patients suffering from an authorized list of severe
or chronic illness. Cannabis has been used to treat a variety of conditions: as an analgesic to
treat neuropathic pain, as antinauseant and antiemetic action in treatment of patients receiving
radiation therapy or chemotherapy for AIDS or cancer an antiemetic, as an appetite stimulant
in patients with anorexia and wasting syndromes, the reduction of intraocular pressure in the
treatment of glaucoma, anticonvulsant action as an adjuvant therapy for epilepsy, and as an aid
in the relief muscle spasticity in patient with muscular sclerosis (9).
                                     ACKNOWLEDGMENTS
We thank the generous support of Bratty Family Fund, Michael and Karyn Goldstein Cancer
Research Fund, Joey and Mary Furfari Cancer Research Fund, Pulenzas Cancer Research Fund,
Joseph and Silvana Melara Cancer Research Fund, and Ofelia Cancer Research Fund.
                                            REFERENCES
[1]    Mikuriya TH. Marijuana in medicine: past, present and future. California Med 1969;110:34-40.
[2]    Schultes RE. Random thoughts and queries on the botany of cannabis. In: Joyce CRB, Curry SH, eds.
       The botany and chemistry of cannabis. London: J & A Churchill, 1970:11-38.
[3]    Commission of Inquiry into the Non-Medical Use of Drugs (Le Dain Commission). Cannabis. Ottawa:
       Information Canada, 1972:11-25,125-126.
[4]    Fairbairn JW, Liebman JA, Rowan MG. The stability of cannabis and its preparations on storage. J
       Pharm Pharmacol 1976;28:1-7.
[5]    Rilery D. Drugs and drug policy in Canada: a brief review and commentary. Canadian Foundation
       for Drug Policy & International Harm Reduction Association 1998 URL: http://www.parl.gc.ca/
       Content/SEN/Committee/362/ille/rep/rep-nov98-e.htm.
[6]    Giffen J, Endicott S, Lambert S. Panic and indifference – the politics of Canada’s drug laws. Ottawa:
       Canadian Centre Substance Abuse, 1991.
[7]    A timeline of some significant events in the history of marijuana in Canada. Canadian Press 2014 URL:
       http://cponline.thecanadianpress.com/graphics/2014/medical-marijuana-timeline/
[8]    Belle-Isle L, Walsh Z, Callaway R, Lucas P, Capler R, Kay R, et al. Barriers to access for Canadians
       who use cannabis for therapeutic purposes. Int J Drug Policy 2014;25(4):691-9.
[9]    Kalant H. Medicinal use of cannabis: history and current status. Pain Res Manag 2001;6(2):80-91.
[10]   Health Canada. Harper government announces new medical marihuana regulations. Canada Gazette
       2013 URL: http://www.hc-sc.gc.ca/ahc-asc/media/nr-cp/_2013/2013-79-eng.php.
[11]   Le Dain Commission. Cannabis: A report of the commission in the inquiry into the non-medical use of
       drugs. Ottawa, ON: Information Canada, 1972.
[12]   Fischer B, Kuganesan S, Room R. Medical marijuana programs: Implications for cannabis control
       policy – observations from Canada. Int J Drug Policy 2015;26(1):15-9.
[13]   Liberal Party of Canada. Real change: A new plan for a strong middle class 2015 URL:
       https://www.liberal.ca/files/2015/10/New-plan-for-a-strong-middle-class.pdf.
[14]   Kilmer B. Policy designs for cannabis legalization: starting with the eight Ps. Am J Drug Alcohol Abuse
       2014;40(4):259-61.
[15]   Van Gerpen S, Vik T, Soundy TJ. Medicinal and recreational marijuana: what are the risks? S D Med
       2015;58-62.
[16]   Meier MH, Caspi A, Ambler A, Harrington H, Houts R, Keefe RS, et al. Persistent cannabis users show
       neuropsychological decline from childhood to midlife. Proc Natl Acad Sci USA 2012;109(40):E2657-
       64.
[17]   Health Canada. Information for health care professionals: cannabis (marihuana, marijuana) and the
       cannabinoids. URL http://www.hc-sc.gc.ca/dhp-mps/marihuana/med/infoprof-eng.php.
[18]   Adda J, McConnell B, Rasul I. Crime and the decriminalization of cannabis: evidence from a localized
       policing experiment 2010 URL: http://www.iza.org/conference_files/riskonomics2011/mcconnell_
       b6110.pdf.
[19]   Hall W, Weier M. Assessing the public health impacts of legalizing recreational cannabis use in the
       USA. Clin Pharmacol Ther 2015;97(6):607-15.
[20]   Anderson DM, Rees DI, Sabia JJ. Medical marijuana laws and suicides by gender and age. Am J Public
       Health 2014;104(12):2369-76.
                                Canada and medical marijuana                                      225
[21]   Rylander M, Valdez C, Nussbaum AM. Does the legalization of medical marijuana increase completed
       suicide? Am J Drug Alcohol Abuse 2014;40(4):269-73.
[22]   Collen M. Prescribing cannabis for harm reduction. Harm Reduct J 2012;9:1.
In: Cannabis: Medical Aspects                                            ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.                   © 2017 Nova Science Publishers, Inc.
Chapter 20
      Palliative medicine focuses on relieving the symptoms and supporting the needs of patients
      and families faced with terminal illness. It embraces a biopsychosocial model of care for
      patients and their loved ones. Attitudes towards use of medical cannabis are becoming
      more liberal among those in the palliative care community as well, and for good reason.
      Marijuana is relatively safe in comparison with prescription narcotics, both in terms of the
      individual patient’s health and public health as there is less damage done by diversion.
      Consequently, treatment with medical cannabis presents an opportunity for patient
      autonomy, for controlling an aspect of their medication. However, additional research is
      required into the substance’s safety and efficacy.
                                          INTRODUCTION
We are called upon as physicians to provide comfort always, but perhaps at no other time in a
person's’ life is managing their comfort as critical, and their vulnerability more evident, than
when they are living with a debilitating illness.
    Palliative medicine focuses on relieving the symptoms and supporting the needs of patients
and families faced with terminal illness. It embraces a biopsychosocial model of care for
    Correspondence: Marissa Slaven, MD, Faculty of Health Sciences, McMaster University, Hamilton, Ontario,
      Canada. E-mail: slavenm@hhsc.ca.
228                       Noah Spencer, Erynn Shaw and Marissa Slaven
patients and their loved ones. In this short communication, we consider the impacts of medical
cannabis on the physical, psychological and social aspects of patient care.
they can discuss it further with you.” The maintenance of the clinician-patient relationship is
important for many reasons, not least of which is that if patients are not directed to appropriate
resources, then they may still seek cannabis for medical purposes in places other than the state-
sanctioned facilities. Products from local suppliers are far more prone to contaminants and
place patients at unnecessary risk. Further, in this scenario, patients may not disclose to their
physicians that they are using cannabis, which may impact their clinical care.
                                              CONCLUSION
A survey conducted in 2001 on physicians’ views toward prescribing cannabis revealed that
only a third of physicians polled would prescribe cannabis if it were legal (5). In a 2014
WebMD survey of 1,554 physicians in more than 12 specialties, 69% believed it can help with
certain treatments and conditions. Hematologists and oncologists were most likely (82%) to
agree that cannabis delivers health benefits. Attitudes towards use of medical cannabis are
becoming more liberal among those in the palliative care community as well, and for good
reason. Marijuana is relatively safe in comparison with prescription narcotics, both in terms of
the individual patient’s health and public health as there is less damage done by diversion.
Consequently, treatment with medical cannabis presents an opportunity for patient autonomy,
for controlling an aspect of their medication.
     However, additional research is required into the substance’s safety and efficacy. Research
into proper dosages and intake methods would be especially beneficial to doctors and patients.
For this research to occur and for other reasons, it is necessary that the stigma against cannabis
use be broken down. Doctors, patients, and the general population must come to understand
that cannabis has a place in healthcare institutions like hospitals and laboratories. If this
research does occur and reflects positively on medical cannabis, it is our hope that this product
will begin to see increased use where appropriate and also that it will be covered by provincial
and/or private insurance plans. Finally, patients must be reminded that the cannabis they buy
from local suppliers is not safe, and that they should seek medical avenues to safely obtain a
quality-controlled product.
     This modern version of the Hippocratic Oath includes the following statement:
           I will remember that there is art to medicine as well as science, and that warmth,
            sympathy, and understanding may outweigh the surgeon's knife or the chemist's drug.
           In using medical cannabis to help bring comfort to palliative patients we call upon
            healthcare professionals to embrace both the science and the art of medicine.
                                              REFERENCES
[1]       Grant I, Atkinson JH, Gouaux B, Wilsey B. Medical marijuana: clearing away the smoke. Open Neurol
          J 2012;6:18-25.
[2]       Grotenhermen F, Muller-Vahl K. The therapeutic potential of cannabis and cannabinoids. Dtsch Arztebl
          Int 2012;109(29-30):495-501.
[3]       National Cancer Institute. Cannabis and cannabinoids - health professional version. National Cancer
          Institute. URL: http://www.cancer.gov/about-cancer/treatment/cam/hp/cannabis-pdq.
                            Medical cannabis and palliative care                                 231
Chapter 21
Edward Chow, MBBS, MSc, PhD, FRCPC is Professor of Radiation Oncology at the
University of Toronto in Canada. He is the chair of the Rapid Response Radiotherapy Program
and Bone Metastases Site Group at the Odette Cancer Centre, Sunnybrook Health Sciences
Centre, Toronto, Canada and also a senior scientist at the Sunnybrook Research Institute. He
has published in the area of palliative radiotherapy and end of life care issues. E-mail:
Edward.Chow@sunnybrook.ca
Hatim A Omar, MD, FAAP, Professor of Pediatrics and Obstetrics and Gynecology;
Professor of Family Studies; and Chief of the Division of Adolescent Medicine, Department of
Pediatrics, University of Kentucky, Lexington. He is the holder of the Children’s Miracle
Network Endowed Chair in Pediatrics. Dr. Omar has completed residency training in obstetrics
and gynecology as well as pediatrics. He has also completed fellowships in vascular physiology
and adolescent medicine. Dr. Omar is the founder and chairman of the Stop Youth Suicide
Cmpaign. He is the recipient of the Commonwealth of Kentucky Governor’s Award for
community service and volunteerism in 2000, Kentucky Teen Pregnancy Coalition Award for
outstanding service 2002, Awards for suicide prevention from the Ohio Valley Society for
Adolescent Medicine and Kentucky Pediatric Society in 2005 and 2007, Sexual Abuse
Awareness Month Award for his work with sexual abuse victims from the Kentucky
Association of Sexual Assault Professionals in 2007, Special Achievement Award from the
American Academy of Pediatrics 2007 and the Founders of Adolescent Medicine Award from
236                      Blair Henry, Arnav Agarwal, Edward Chow et al.
the AAP in 2007. He is well known internationally with numerous publications in child health,
public health, pediatrics, adolescent medicine, pediatric and adolescent gynecology. E-mail:
haomar2@uky.edu
Joav Merrick, MD, MMedSci, DMSc, born and educated in Denmark is professor of
pediatrics, child health and human development, Division of Pediatrics, Hadassah Hebrew
University Medical Center, Mt Scopus Campus, Jerusalem, Israel and Kentucky Children’s
Hospital, University of Kentucky, Lexington, Kentucky United States and professor of public
health at the Center for Healthy Development, School of Public Health, Georgia State
University, Atlanta, United States, the medical director of the Health Services, Division for
Intellectual and Developmental Disabilities, Ministry of Social Affairs and Social Services,
Jerusalem, the founder and director of the National Institute of Child Health and Human
Development in Israel. Numerous publications in the field of pediatrics, child health and human
development, rehabilitation, intellectual disability, disability, health, welfare, abuse, advocacy,
quality of life and prevention. Received the Peter Sabroe Child Award for outstanding work on
behalf of Danish Children in 1985 and the International LEGO-Prize (“The Children’s Nobel
Prize”) for an extraordinary contribution towards improvement in child welfare and well-being
in 1987. E-mail: jmerrick@zahav.net.il
In: Cannabis: Medical Aspects                                     ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.            © 2017 Nova Science Publishers, Inc.
Chapter 22
The Odette Cancer Centre, the comprehensive cancer program of Sunnybrook Health Sciences
Centre is a leading regional cancer centre in Toronto, Ontario, Canada. It is the sixth largest
cancer centre in North America in terms of number of new cancer patients seen per year. The
Department of Radiation Oncology at Sunnybrook is an academic unit fully affiliated with the
University of Toronto. Palliative radiotherapy is one of the key research foci in the Department
of Radiation Oncology. The Rapid Response Radiotherapy Program (RRRP) is a specialized
clinic designed to provide timely palliative radiotherapy. The RRRP was developed in 1996,
and approximately 500-600 patients are seen in one year. This program aims to improve the
quality of life of palliative cancer patients, while decreasing wait time and allowing for same
day treatment.
Contact
Professor Edward Chow, MBBS, PhD, FRCPC, Department of Radiation Oncology, Odette
Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, Ontario,
Canada M4N 3M5. E-mail: Edward.Chow@sunnybrook.ca
In: Cannabis: Medical Aspects                                      ISBN: 978-1-53610-510-0
Editors: B. Henry, A. Agarwal, E. Chow et al.             © 2017 Nova Science Publishers, Inc.
Chapter 23
The National Institute of Child Health and Human Development (NICHD) in Israel was
established in 1998 as a virtual institute under the auspices of the Medical Director, Ministry
of Social Affairs and Social Services in order to function as the research arm for the Office of
the Medical Director. In 1998 the National Council for Child Health and Pediatrics, Ministry
of Health and in 1999 the Director General and Deputy Director General of the Ministry of
Health endorsed the establishment of the NICHD.
Mission
The mission of a National Institute for Child Health and Human Development in Israel is to
provide an academic focal point for the scholarly interdisciplinary study of child life, health,
public health, welfare, disability, rehabilitation, intellectual disability and related aspects of
human development. This mission includes research, teaching, clinical work, information and
public service activities in the field of child health and human development.
Over the years many activities became focused in the south of Israel due to collaboration with
various professionals at the Faculty of Health Sciences (FOHS) at the Ben Gurion University
of the Negev (BGU). Since 2000 an affiliation with the Zusman Child Development Center at
the Pediatric Division of Soroka University Medical Center has resulted in collaboration around
the establishment of the Down Syndrome Clinic at that center. In 2002 a full course on
“Disability” was established at the Recanati School for Allied Professions in the Community,
FOHS, BGU and in 2005 collaboration was started with the Primary Care Unit of the faculty
and disability became part of the master of public health course on “Children and society”. In
the academic year 2005-2006 a one semester course on “Aging with disability” was started as
part of the master of science program in gerontology in our collaboration with the Center for
Multidisciplinary Research in Aging. In 2010 collaborations with the Division of Pediatrics,
Hadassah Hebrew University Medical Center, Jerusalem, Israel around the National Down
240                     Blair Henry, Arnav Agarwal, Edward Chow et al.
Syndrome Center and teaching students and residents about intellectual and developmental
disabilities as part of their training at this campus.
Research activities
The affiliated staff have over the years published work from projects and research activities in
this national and international collaboration. In the year 2000 the International Journal of
Adolescent Medicine and Health and in 2005 the International Journal on Disability and Human
Development of De Gruyter Publishing House (Berlin and New York) were affiliated with the
National Institute of Child Health and Human Development. From 2008 also the International
Journal of Child Health and Human Development (Nova Science, New York), the International
Journal of Child and Adolescent Health (Nova Science) and the Journal of Pain Management
(Nova Science) affiliated and from 2009 the International Public Health Journal (Nova Science)
and Journal of Alternative Medicine Research (Nova Science). All peer-reviewed international
journals.
National collaborations
Nationally the NICHD works in collaboration with the Faculty of Health Sciences, Ben Gurion
University of the Negev; Department of Physical Therapy, Sackler School of Medicine, Tel
Aviv University; Autism Center, Assaf HaRofeh Medical Center; National Rett and PKU
Centers at Chaim Sheba Medical Center, Tel HaShomer; Department of Physiotherapy, Haifa
University; Department of Education, Bar Ilan University, Ramat Gan, Faculty of Social
Sciences and Health Sciences; College of Judea and Samaria in Ariel and in 2011 affiliation
with Center for Pediatric Chronic Diseases and National Center for Down Syndrome,
Department of Pediatrics, Hadassah Hebrew University Medical Center, Mount Scopus
Campus, Jerusalem.
International collaborations
Internationally with the Department of Disability and Human Development, College of Applied
Health Sciences, University of Illinois at Chicago; Strong Center for Developmental
Disabilities, Golisano Children's Hospital at Strong, University of Rochester School of
Medicine and Dentistry, New York; Centre on Intellectual Disabilities, University of Albany,
New York; Centre for Chronic Disease Prevention and Control, Health Canada, Ottawa;
Chandler Medical Center and Children’s Hospital, Kentucky Children’s Hospital, Section of
Adolescent Medicine, University of Kentucky, Lexington; Chronic Disease Prevention and
Control Research Center, Baylor College of Medicine, Houston, Texas; Division of
Neuroscience, Department of Psychiatry, Columbia University, New York; Institute for the
Study of Disadvantage and Disability, Atlanta; Center for Autism and Related Disorders,
Department Psychiatry, Children’s Hospital Boston, Boston; Department of Pediatric and
Adolescent Medicine, Western Michigan University Homer Stryker MD School of Medicine,
Kalamazoo, Michigan, United States; Department of Paediatrics, Child Health and Adolescent
Medicine, Children's Hospital at Westmead, Westmead, Australia; International Centre for the
      About the National Institute of Child Health and Human Development, Israel           241
Study of Occupational and Mental Health, Düsseldorf, Germany; Centre for Advanced Studies
in Nursing, Department of General Practice and Primary Care, University of Aberdeen,
Aberdeen, United Kingdom; Quality of Life Research Center, Copenhagen, Denmark; Nordic
School of Public Health, Gottenburg, Sweden, Scandinavian Institute of Quality of Working
Life, Oslo, Norway; The Department of Applied Social Sciences (APSS) of The Hong Kong
Polytechnic University Hong Kong.
Targets
Our focus is on research, international collaborations, clinical work, teaching and policy in
health, disability and human development and to establish the NICHD as a permanent institute
in Israel in order to conduct model research and together with the four university schools of
public health/medicine in Israel establish a national master and doctoral program in disability
and human development at the institute to secure the next generation of professionals working
in this often non-prestigious/low-status field of work.
Contact
Chapter 24
Health and human development is a book series with publications from a multidisciplinary
group of researchers, practitioners and clinicians for an international professional forum
interested in the broad spectrum of health and human development. Books already published:
   Wernik U. Chance action and therapy. The playful way of changing. New York: Nova
    Science, 2010.
   Omar HA, Greydanus DE, Patel DR, Merrick J, eds. Adolescence and chronic illness. A
    public health concern. New York: Nova Science, 2010.
   Patel DR, Greydanus DE, Omar HA, Merrick J, eds. Adolescence and sports. New York:
    Nova Science, 2010.
   Shek DTL, Ma HK, Merrick J, eds. Positive youth development: Evaluation and future
    directions in a Chinese context. New York: Nova Science, 2010.
   Shek DTL, Ma HK, Merrick J, eds. Positive youth development: Implementation of a
    youth program in a Chinese context. New York: Nova Science, 2010.
   Omar HA, Greydanus DE, Tsitsika AK, Patel DR, Merrick J, eds. Pediatric and
    adolescent sexuality and gynecology: Principles for the primary care clinician. New
    York: Nova Science, 2010.
   Chow E, Merrick J, eds. Advanced cancer. Pain and quality of life. New York: Nova
    Science, 2010.
   Latzer Y, Merrick, J, Stein D, eds. Understanding eating disorders. Integrating culture,
    psychology and biology. New York: Nova Science, 2010.
   Sahgal A, Chow E, Merrick J, eds. Bone and brain metastases: Advances in research and
    treatment. New York: Nova Science, 2010.
   Postolache TT, Merrick J, eds. Environment, mood disorders and suicide. New York:
    Nova Science, 2010.
   Maharajh HD, Merrick J, eds. Social and cultural psychiatry experience from the
    Caribbean Region. New York: Nova Science, 2010.
   Mirsky J. Narratives and meanings of migration. New York: Nova Science, 2010.
   Harvey PW. Self-management and the health care consumer. New York: Nova Science,
    2011.
   Ventegodt S, Merrick J. Sexology from a holistic point of view. New York: Nova
    Science, 2011.
   Ventegodt S, Merrick J. Principles of holistic psychiatry: A textbook on holistic
    medicine for mental disorders. New York: Nova Science, 2011.
   Greydanus DE, Calles Jr JL, Patel DR, Nazeer A, Merrick J, eds. Clinical aspects of
    psychopharmacology in childhood and adolescence. New York: Nova Science, 2011.
   Bell E, Seidel BM, Merrick J, eds. Climate change and rural child health. New York:
    Nova Science, 2011.
   Bell E, Zimitat C, Merrick J, eds. Rural medical education: Practical strategies. New
    York: Nova Science, 2011.
   Latzer Y, Tzischinsky. The dance of sleeping and eating among adolescents: Normal
    and pathological perspectives. New York: Nova Science, 2011.
   Deshmukh VD. The astonishing brain and holistic consciousness: Neuroscience and
    Vedanta perspectives. New York: Nova Science, 2011.
   Bell E, Westert GP, Merrick J, eds. Translational research for primary healthcare. New
    York: Nova Science, 2011.
   Shek DTL, Sun RCF, Merrick J, eds. Drug abuse in Hong Kong: Development and
    evaluation of a prevention program. New York: Nova Science, 2011.
   Ventegodt S, Hermansen TD, Merrick J. Human Development: Biology from a holistic
    point of view. New York: Nova Science, 2011.
   Ventegodt S, Merrick J. Our search for meaning in life. New York: Nova Science, 2011.
   Caron RM, Merrick J, eds. Building community capacity: Minority and immigrant
    populations. New York: Nova Science, 2012.
            About the Book Series “Health and Human Development”                     245
 Klein H, Merrick J, eds. Human immunodeficiency virus (HIV) research: Social science
  aspects. New York: Nova Science, 2012.
 Lutzker JR, Merrick J, eds. Applied public health: Examining multifaceted Social or
  ecological problems and child maltreatment. New York: Nova Science, 2012.
 Chemtob D, Merrick J, eds. AIDS and tuberculosis: Public health aspects. New York:
  Nova Science, 2012.
 Ventegodt S, Merrick J. Textbook on evidence-based holistic mind-body medicine:
  Basic principles of healing in traditional Hippocratic medicine. New York: Nova
  Science, 2012.
 Ventegodt S, Merrick J. Textbook on evidence-based holistic mind-body medicine:
  Holistic practice of traditional Hippocratic medicine. New York: Nova Science, 2012.
 Ventegodt S, Merrick J. Textbook on evidence-based holistic mind-body medicine:
  Healing the mind in traditional Hippocratic medicine. New York: Nova Science, 2012.
 Ventegodt S, Merrick J. Textbook on evidence-based holistic mind-body medicine:
  Sexology and traditional Hippocratic medicine. New York: Nova Science, 2012.
 Ventegodt S, Merrick J. Textbook on evidence-based holistic mind-body medicine:
  Research, philosophy, economy and politics of traditional Hippocratic medicine. New
  York: Nova Science, 2012.
 Caron RM, Merrick J, eds. Building community capacity: Skills and principles. New
  York: Nova Science, 2012.
 Lemal M, Merrick J, eds. Health risk communication. New York: Nova Science, 2012.
 Ventegodt S, Merrick J. Textbook on evidence-based holistic mind-body medicine:
  Basic philosophy and ethics of traditional Hippocratic medicine. New York: Nova
  Science, 2013.
 Caron RM, Merrick J, eds. Building community capacity: Case examples from around
  the world. New York: Nova Science, 2013.
 Steele RE. Managed care in a public setting. New York: Nova Science, 2013.
 Srabstein JC, Merrick J, eds. Bullying: A public health concern. New York: Nova
  Science, 2013.
 Pulenzas N, Lechner B, Thavarajah N, Chow E, Merrick J, eds. Advanced cancer:
  Managing symptoms and quality of life. New York: Nova Science, 2013.
 Stein D, Latzer Y, eds. Treatment and recovery of eating disorders. New York: Nova
  Science, 2013.
 Sun J, Buys N, Merrick J. Health promotion: Community singing as a vehicle to promote
  health. New York: Nova Science, 2013.
 Pulenzas N, Lechner B, Thavarajah N, Chow E, Merrick J, eds. Advanced cancer:
  Managing symptoms and quality of life. New York: Nova Science, 2013.
 Sun J, Buys N, Merrick J. Health promotion: Strengthening positive health and
  preventing disease. New York: Nova Science, 2013.
 Merrick J, Israeli S, eds. Food, nutrition and eating behavior. New York: Nova Science,
  2013.
 Shahtahmasebi S, Merrick J. Suicide from a public health perspective.
  New York: Nova Science, 2014.
 Merrick J, Tenenbaum A, eds. Public health concern: Smoking, alcohol and substance
  use. New York: Nova Science, 2014.
 Merrick J, Aspler S, Morad M, eds. Mental health from an international perspective.
  New York: Nova Science, 2014.
 Merrick J, ed. India: Health and human development aspects. New York: Nova Science,
  2014.
246                     Blair Henry, Arnav Agarwal, Edward Chow et al.
Contact
Food and Drug Administration (FDA), 5, 7, 46, 54,      guidelines, 101, 105, 110, 121, 124, 141, 151, 179,
   108, 147, 148, 149, 151, 152, 182, 184                 180, 185, 189, 190, 196, 197, 199, 206
food intake, 114, 119, 120, 123, 127                   guilt, 101
formaldehyde, 30                                       gynecomastia, 208
formation, 81, 192
fractures, 246
France, 4, 30, 182, 218
                                                                                  H
free radicals, 78
                                                       hair, 36, 53, 68, 70
freedom, 18
                                                       half-life, 67
frontal cortex, 57
                                                       hallucinations, 35, 37, 44
functional MRI, 44
                                                       happiness, 18
fusion, 80
                                                       harmful effects, 150, 204
                                                       hazards, 55, 142
                          G                            head and neck cancer, 93, 128, 246
                                                       headache, 7, 136, 139, 180, 183
GABA, 67, 80, 106, 192                                 health care, 4, 25, 81, 92, 101, 102, 144, 193, 197,
galactorrhea, 208                                         206, 224, 244
gastric mucosa, 80                                     health care professionals, 25, 81, 102, 144, 197, 224
gastroesophageal reflux, 36                            health condition, 27, 28, 65, 66, 147, 179
gastrointestinal tract, 24, 32, 33, 37, 140            health effects, 52, 181, 184, 209, 223, 243
gender differences, 109                                health problems, 27, 28, 185, 218
gene expression, 23                                    health risks, 14, 185
gene promoter, 23                                      heart disease, 9, 12, 39
generalizability, 91, 141                              heart failure, 9
genetic factors, 31, 45                                heart rate, 39, 40, 228
genus, 72, 99, 179                                     height, 97
Georgia, xvii, 27, 107, 236                            hematology, 180, 185
Germany, 4, 7, 30, 161, 182, 241                       hematopoietic system, 99
germination, 35                                        hemoglobin, 228
gerontology, 239                                       hemorrhoids, 86
gestational diabetes, 9                                hemp, 16, 19, 20, 25, 28, 31, 79, 80, 121, 125, 147,
glaucoma, 34, 148, 149, 201, 222                          197, 221
Glioblastoma, 9                                        hepatitis, 148, 149
global markets, 15                                     herbal medicine, 15, 77
globalization, 246                                     heroin, 10, 11, 28, 41, 42, 44, 59
glucose, 208                                           heterogeneity, 40
glutamate, 47, 67, 74, 80, 192                         high school, 29, 181, 207, 208, 212, 214, 216, 218,
glycerol, 28, 32                                          219
glycol, 23                                             higher education, 246
glycoproteins, 77                                      high-risk populations, 184
goal attainment, 220                                   hippocampus, 32, 33, 41, 42, 47, 57, 66, 106, 197
gonorrhea, 19                                          historical overview, 14
gout, 7, 18, 86                                        history, xvii, xix, 3, 14, 15, 17, 24, 39, 40, 42, 45, 72,
governance, 215                                           88, 102, 114, 130, 137, 140, 143, 190, 221, 224
Greece, 10, 30                                         HIV, 7, 34, 49, 82, 105, 109, 115, 123, 124, 126,
Greeks, 86, 92                                            127, 128, 134, 135, 137, 138, 141, 143, 145, 149,
growth, 4, 8, 33, 53, 59, 114, 208                        156, 158, 159, 175, 178, 180, 190, 194, 196, 201,
guardian, 18, 182                                         245
guessing, 141                                          holistic medicine, 244
guidance, xix, 26, 96, 199, 205, 206, 219              Hong Kong, 241, 244
                                                       hormone, 47, 208
256                                                      Index
hospitalization, 39, 212, 213, 216                        inflammatory bowel disease, 34, 49, 52, 195, 198
host, 208, 209                                            inflammatory disease, 77
House, 177, 240                                           inflammatory responses, 106
human body, 10, 71, 99                                    influenza, 8
human brain, 58                                           informed consent, xix
human development, xv, 236, 239, 241, 243, 245            ingestion, 23, 24, 35, 36, 39, 65, 67, 75, 76, 109,
human immunodeficiency virus, 148                             120, 121, 140, 146, 147, 155, 157, 194, 208
human subjects, 44                                        ingredients, 3
Hungary, 15, 30                                           inheritance, 20, 25
hydrocarbons, 77                                          inhibition, 5, 8, 9, 31, 33, 51, 66, 74, 80, 106, 130,
hydrogen, 78                                                  192, 197
hyperactivity, 40, 41, 51                                 inhibitor, 6, 7, 8, 9, 48
hyperemesis, 37, 54, 102, 103, 181, 209, 210              initiation, 31, 44, 51, 59, 60, 191, 198, 205, 218
hyperinflation, 38                                        injections, 197
hypertension, 8, 9, 33                                    injuries, 9, 10, 40, 55, 97
hypodermic needles, xvii, 11                              injury, xii, 8, 34, 38, 39, 50, 96, 97
hypothalamus, 43, 106, 114, 209                           insects, 77
hypothesis, 57                                            insomnia, 46, 76, 86, 193, 209, 228
                                                          institutions, 200, 205, 230
                                                          insulin, 9
                          I                               insulin resistance, 9
                                                          integration, 17, 114, 243
IBD, 49, 195
                                                          interference, 40
Iceland, 30
                                                          International Olympic Committee, 40
ideals, 50, 192
                                                          interneurons, 32
identification, 20, 36, 43, 48, 213, 219
                                                          intervention, 50, 52, 59, 120, 121, 122, 124, 125,
idiosyncratic, 36, 217
                                                              128, 129, 131, 135, 137, 138, 140, 141, 143, 144,
illicit drug use, 50, 219
                                                              145, 155, 156, 212, 213, 214, 219
illicit substances, 184
                                                          intoxication, 35, 40, 42, 46, 53, 78, 181
immune function, 15, 99
                                                          intraocular pressure, 31, 34, 222
immune system, 33, 67, 74, 81, 106, 140
                                                          intravenous fluids, 37
immunity, 14
                                                          ion channels, 67, 77, 78
immunodeficiency, 245
                                                          ionization, 151
immunosuppression, 81
                                                          Iran, 25, 29, 51, 70, 81, 127
improvements, 95, 109
                                                          Ireland, 4, 30
impulsivity, 40
                                                          iris, 197
impurities, 35
                                                          irradiation, 78
in vitro, 4, 69, 78, 81, 229
                                                          irritability, 42, 181, 209
in vivo, 58, 70, 74, 80
                                                          irritable bowel syndrome, 8, 49
incidence, 31, 123, 174, 181
                                                          isolation, 7, 11, 43, 114
India, 10, 15, 18, 19, 25, 92, 182, 245
                                                          Israel, xv, xvii, xviii, 27, 33, 111, 116, 179, 182, 236,
individual differences, 101
                                                              239, 241, 246
individuality, 4
                                                          issues, 11, 15, 121, 138, 184, 199, 200, 203, 208,
individuals, 19, 22, 24, 34, 44, 45, 47, 49, 109, 135,
                                                              209, 212, 213, 215, 223, 228, 229, 235
    137, 139, 141, 150, 153, 174, 181, 182, 193, 200,
                                                          Italy, 30, 182
    201, 202, 203, 208, 211, 223
inducer, 6
induction, 8                                                                         J
industry, 14, 82, 99, 101, 102
infection, 38, 127, 128                                   Jamaica, 182
inflammation, 8, 9, 10, 11, 19, 34, 54, 97, 99, 130,      Japan, 28
    140                                                   joints, 38
                                                  Index                                                       257
K M
keratosis, 6                                              machinery, 80
kidney, 8, 10                                             macrophages, 67
                                                          major depression, 56, 57
                                                          major depressive disorder, 40
                          L                               majority, 42, 68, 86, 87, 88, 91, 92, 101, 102, 108,
                                                            109, 120, 143, 156, 174, 176, 182, 213, 216, 223,
labeling, 109, 147, 151
                                                            228
landscape, 53, 199, 200
                                                          malaria, 18, 77
lateral sclerosis, 48
                                                          malignancy, 88
Latvia, 30
                                                          malignant tumors, 180
law enforcement, 42, 101
                                                          management, xix, 3, 10, 14, 15, 27, 32, 33, 34, 36,
laws, 28, 103, 111, 145, 179, 182, 183, 186, 196,
                                                            41, 42, 44, 45, 46, 47, 48, 51, 53, 59, 85, 86, 92,
    203, 206, 224
                                                            95, 96, 97, 108, 109, 110, 113, 114, 129, 132,
lean body mass, 114, 124
                                                            140, 149, 177, 180, 189, 191, 197, 199, 200, 201,
learning, 100, 106, 212, 213, 246
                                                            219, 228, 229, 244, 246
learning difficulties, 213
                                                          manipulation, 32
legality, 85, 86
                                                          manufacturing, 35
legislation, 19, 100, 105, 109, 110, 179, 180, 182,
                                                          Marihuana, 25, 29, 49, 81, 90, 92, 100, 102, 103,
    184, 186, 189, 199, 200, 202, 204, 205, 206
                                                            111, 125, 127, 131, 144, 145, 189, 190, 191, 192,
lesions, 37, 38, 181
                                                            193, 194, 195, 196, 197, 199, 200, 201, 206, 210,
leukemia, 9, 54, 180
                                                            221, 222, 224
liberty, 206, 229
                                                          Marihuana for Medicinal Purposes Regulations
libido, 208
                                                            (MMPR), 100, 106, 125, 131, 144, 189, 190, 193,
lifetime, 29, 195
                                                            199, 200, 222, 223
light, 8, 12, 106, 194, 209
                                                          marijuana, xiii, xiv, xvii, xviii, xix, 16, 25, 27, 28,
Likert scale, 173
                                                            29, 30, 31, 33, 37, 38, 39, 40, 41, 42, 43, 44, 46,
limbic system, 32, 33
                                                            47, 49, 50, 51, 52, 53, 54, 55, 56, 60, 61, 65, 70,
lipid metabolism, 9
                                                            79, 80, 81, 92, 99, 100, 102, 103, 111, 113, 114,
lipids, 78
                                                            116, 120, 121, 124, 125, 127, 128, 130, 132, 144,
liquid chromatography, 35, 53, 60, 68, 152
                                                            145, 146, 147, 148, 149, 150, 151, 152, 153, 156,
lithium, 48
                                                            178, 179, 180, 181, 182, 183, 184, 185, 186, 195,
Lithuania, 30
                                                            197, 200, 202, 206, 207, 208, 209, 210, 211, 212,
liver disease, 9, 12, 16, 102, 103
                                                            213, 214, 215, 216, 217, 218, 219, 221, 222, 223,
liver transplant, 196, 198
                                                            224, 225, 230, 231
local anesthetic, 37
                                                          marketing, 107
locus, 22, 199, 200
                                                          mass spectrometry, 36, 53, 68, 70
longitudinal study, 124, 218
                                                          mastectomy, 190
loss of appetite, 49, 97
                                                          materials, 142
low risk, 14, 65, 141, 207
                                                          matter, xii, 38, 41, 55, 150, 151, 181, 206
LSD, 30
                                                          measurement, 12
lumbar spine, 96
                                                          media, 27, 28, 145, 224, 229
lung cancer, 8, 9, 39, 55, 85, 87, 128, 208, 228
                                                          median, 90, 173
lung disease, 210
                                                          medical, xiii, xiv, xvii, xviii, xix, 3, 10, 11, 12, 14,
lung function, 54
                                                            16, 17, 18, 19, 21, 23, 24, 25, 27, 28, 29, 33, 41,
luteinizing hormone, 35
                                                            48, 49, 50, 61, 69, 81, 85, 86, 87, 90, 91, 92, 95,
lymphocytes, 114
                                                            97, 99, 100, 101, 102, 103, 105, 106, 107, 108,
258                                                        Index
  109, 110, 111, 114, 119, 120, 121, 127, 128, 129,         metabolizing, 48
  130,131, 136, 139, 141, 144, 145, 146, 147, 148,          metastasis, 33
  149, 150, 151, 152, 153, 179, 180, 181, 182, 183,         methadone, 191
  184, 185, 186, 189, 190, 191, 192, 193, 194, 195,         methamphetamine, 28, 31, 45, 59
  196, 198, 199, 200, 201, 202, 206, 209, 221, 222,         methodology, 24, 115, 125, 131
  223, 224, 225, 227, 228, 229, 230, 231, 235, 236,         methyl groups, 78
  244                                                       methylation, 58
medical cannabis, xiii, xiv, xvii, xviii, xix, 14, 16,      metropolitan areas, 152
  17, 23, 28, 33, 48, 49, 60, 81, 85, 86, 87, 90, 91,       Mexico, 19, 107
  92, 95, 97, 99, 100, 101, 102, 103, 105, 106, 107,        mice, 74, 78, 81, 82, 127
  108, 109, 110, 111, 119, 127, 129, 130, 131, 136,         microdialysis, 80
  139, 141, 144, 150, 151, 153, 179, 180, 182, 183,         microsomes, 70
  184, 185, 186,189, 192, 195, 196, 198, 199, 200,          middle class, 224
  206, 223, 227, 228, 229, 230                              Middle East, 4
Medical Marihuana, 189, 190, 191, 192, 193, 194,            migraine headache, 97, 136, 139, 146
  196, 201, 221, 222, 224                                   migraines, 19
Medical marijuana, xiv, xvii, xviii, xix, 16, 49, 50,       migration, 67, 78, 244
  53, 61, 92, 103, 111, 113, 120, 145, 146, 147,            military, 53
  148, 149, 151, 152, 153, 179, 180, 182, 183, 184,         milligrams, 193, 195
  185, 186, 206, 209, 221, 224, 225, 230, 231               mind-body, 245
medication, xvii, 5, 11, 12, 14, 16, 19, 24, 33, 47, 57,    minority groups, 19
  93, 109, 125, 140, 142, 149, 162, 180, 183, 227,          mission, 239
  230                                                       misuse, 16, 51, 179, 185, 186, 199, 202
medicine, xvii, 3, 4, 5, 7, 11, 12, 14, 15, 18, 19, 24,     mixing, 130
  25, 26, 37, 50, 76, 92, 102, 103, 108, 111, 127,          MMS, 25, 81, 127
  177, 221, 222, 223, 224, 227, 228, 229, 230, 235,         models, 52, 56, 131, 161, 219
  241, 243, 245                                             moderators, 59
Mediterranean, xix, 15, 18, 140                             mold, 243
medulla, 106                                                molecular biology, 4
melanoma, 87, 88                                            molecules, 32, 77, 78, 79, 130
mellitus, 9                                                 Montana, 183, 186
membranes, 78                                               mood change, 120
memory, 9, 14, 32, 34, 40, 53, 56, 66, 100, 106, 120,       mood disorder, 109, 140, 244
  181, 217                                                  morbidity, 77, 120, 128, 145
meningitis, 16                                              morphine, xvii, 10, 12, 130, 136, 139, 140, 195, 197,
mental disorder, 51, 56, 59, 219, 244                          198
mental health, 42, 45, 59, 88, 103, 109, 184, 212,          mortality, 3, 16, 34, 36, 39, 77, 114, 120, 128, 145,
  213, 217, 218, 219, 223                                      181, 203, 206, 223
mental health professionals, 217                            motivation, 44, 218
mental illness, 46                                          motor skills, 181
Mesopotamia, 4, 10                                          movement disorders, 49
meta-analysis, 55, 56, 61, 92, 102, 103, 126, 145,          mucosa, 37, 181
  146, 180, 185, 218                                        multidimensional, 45, 59
metabolic changes, 114                                      multiple sclerosis, 7, 33, 34, 48, 49, 52, 100, 103,
metabolic disorders, 197                                       114, 126, 146, 148, 149, 151, 153, 156, 168, 174,
metabolic syndrome, 9                                          175, 177, 178, 180, 182, 197
metabolism, 14, 25, 65, 66, 67, 69, 70, 76, 78, 81,         muscle mass, 124
  101, 121, 127, 142                                        muscle spasms, 19, 86, 228
metabolites, 4, 22, 35, 60, 68, 69, 71, 72, 76, 77, 79,     muscular dystrophy, 8
  137                                                       musculoskeletal, 130, 180
metabolized, 68, 69, 101, 122                               mutant, 22
                                                   Index                                                      259
pain management, 3, 10, 12, 14, 97, 103, 131, 140         pharmacology, xiii, 53, 60, 63, 70, 80, 81, 102, 114,
pain perception, 60, 66, 114, 137, 170                       115, 117, 131
pain tolerance, 137, 139                                  pharmacotherapy, 16, 46, 126, 180
paints, 109                                               pharyngitis, 36
palliate, 127                                             phencyclidine, 28, 30, 42
palliative, xiii, xiv, 49, 61, 85, 86, 90, 91, 92, 93,    phenotype, 20, 25
   120, 123, 125, 205, 227, 228, 229, 230, 235, 237       phenotypes, 22, 25, 80
pancreas, 88                                              Philadelphia, 15, 25
pancreatitis, 36, 49, 54, 195                             physical activity, 36
panic attack, 42, 208                                     physical health, 109, 212
panic disorder, 34                                        physicians, xix, 10, 11, 19, 108, 131, 141, 147, 148,
parallel, 126, 177, 178                                      149, 179, 189, 190, 193, 199, 200, 201, 202, 205,
paranoia, 23, 35, 43, 44, 76, 208                            206, 222, 223, 227, 230
parental involvement, 214, 215                            physiological factors, 67
parent-child relationship, 215, 218                       physiology, 72, 79, 97, 102, 235
parenting, 215                                            Phyto-cannabinoids, 66
parents, 40, 45, 56, 59, 213, 214, 215, 216, 217, 219     pilot study, 26, 57, 111, 118, 120, 127, 145, 177
paresthesias, 190                                         PKU, 240
partial seizure, 52                                       placebo, 52, 81, 107, 108, 117, 118, 119, 120, 123,
participants, 49, 91, 110, 123, 124, 133, 137, 138,          126, 127, 128, 131, 134, 135, 136, 137, 138, 140,
   139, 140, 141, 143, 162, 175, 180                         141, 143, 145, 146, 156, 158, 159, 160, 161, 162,
password, 86                                                 163, 164, 165, 166, 167, 168, 169, 170, 171, 172,
patents, 193, 194                                            173, 174, 175, 176, 177, 178, 180, 190, 194, 195,
pathogenesis, 126                                            196
pathophysiology, 114, 128                                 placenta, 68
pathways, 8, 22, 47, 57, 69, 72, 77, 125, 130             plants, 3, 4, 5, 7, 12, 14, 15, 17, 18, 20, 25, 53, 72,
patient care, 95, 228                                        77, 78, 82, 99, 149, 150, 151, 190
PCP, 30, 42                                               plaque, 8, 9
PCR, 23, 36, 53                                           plasma levels, 76, 140, 142
pediatrician, 219                                         plasticity, 40
peer group, 45, 46                                        playing, 18
peer influence, 219                                       plexus, 177
peptides, 10, 15                                          PM, 69, 185, 217, 218
perforation, 36                                           pneumonia, 181
perinatal, 40                                             pneumothorax, 38
periodontal, 37, 181                                      Poland, 30
periodontal disease, 37, 181                              policy, xiv, 16, 24, 52, 71, 90, 180, 184, 186, 199,
peripheral nervous system, 10, 20, 66, 74                    200, 201, 202, 203, 204, 205, 206, 223, 224, 241
peripheral neuropathy, 16, 177, 180                       policy issues, 199
peripheral vascular disease, 181                          policy makers, 71
peritonitis, 36                                           policy options, 204
permeation, 81                                            politics, 206, 224, 245
permission, xii, 37, 221, 222                             pollen, 17
permit, 119                                               pollination, 20
personal history, 228                                     polycyclic aromatic hydrocarbon, 23, 38
pharmaceutical, 4, 5, 7, 14, 34, 78, 82, 96, 99, 100,     polymorphism, 43
   174, 180                                               polymorphisms, 16
pharmacokinetics, 25, 50, 52, 75, 76, 78, 81, 101,        polyphenols, 77
   109, 127, 128, 145, 194, 197, 198                      polyunsaturated fat, 128
pharmacological treatment, 180, 197                       polyunsaturated fatty acids, 128
                                                   Index                                                      261
population, 40, 44, 51, 55, 90, 91, 92, 120, 123, 124,     psychological dependence, 101
   146, 176, 179, 183, 184, 204, 207, 208, 209, 213,       psychological stress, 45
   215, 230                                                psychological withdrawal, 209
Portugal, 30                                               psychologist, 216
positive correlation, 117, 121                             psychology, 244
positive mood, 47                                          psychopharmacology, 51, 57, 244
positive reinforcement, 142                                psychosis, 8, 35, 37, 43, 44, 45, 47, 48, 50, 58, 69,
positron, 56                                                  102, 103, 161, 208, 212, 219
positron emission tomography, 56                           psychosomatic, 5
posttraumatic stress, 34, 49, 60                           psychotic symptoms, 43, 44
post-traumatic stress disorder (PTSD) , 34, 48, 49,        psychotropic drugs, 53
   96, 105, 201                                            public health, 48, 60, 144, 145, 184, 206, 224, 227,
potassium, 66, 67                                             230, 236, 239, 241, 243, 244, 245, 246
potential benefits, 33                                     public opinion, 109
precipitation, 37                                          public safety, 101, 223
predators, 4, 77                                           public service, 239
prefrontal cortex, 32, 43, 57, 80, 208                     Puerto Rico, 182
preparation, xii, 11, 109, 151                             purity, 222
prescription drug abuse, 223                               pyrolysis, 23, 75, 76, 121
prescription drugs, 11, 31, 46, 184
prevention, 10, 15, 44, 45, 46, 59, 146, 212, 213,
   214, 215, 216, 217, 219, 235, 236, 244
                                                                                      Q
principles, 45, 199, 200, 202, 203, 245
                                                           quality control, 131
problem behavior, 216
                                                           quality of life, 14, 95, 96, 97, 114, 120, 122, 127,
problem solving, 208
                                                             128, 191, 236, 237, 244, 245, 246
procurement, 223
                                                           quality of service, 186
producers, 49, 86, 91, 100, 106, 129, 131, 141, 189,
                                                           query, xviii, 50
   190, 192, 193, 194
                                                           questionnaire, 217
professionals, 213, 215, 216, 217, 228, 229, 230,
   239, 241
prognosis, 86                                                                         R
pro-inflammatory, 74, 114
project, 86                                                radiation, 8, 118, 122, 222
prolactin, 208                                             Radiation, 235, 237
proliferation, xix, 32                                     radiation therapy, 8, 122, 222
promoter, 58                                               radical formation, 5
propagation, 12, 16, 20                                    radiotherapy, 235, 237
prophylaxis, 139                                           rating scale, 138, 146, 162, 180
propylene, 23                                              reaction time, 40, 181
prosocial behavior, 216                                    reactions, 35, 37, 77
prostaglandins, 67, 130                                    reactive oxygen, 78
prostate cancer, 8, 9, 87                                  reality, 142, 208
prostate carcinoma, 34                                     reasoning, 212
protection, 77, 79                                         receptors, 6, 10, 15, 20, 28, 32, 33, 35, 41, 48, 65,
protective factors, 213, 219                                   66, 67, 69, 70, 72, 74, 77, 78, 80, 81, 99, 100,
proteins, 77                                                   106, 114, 121, 130, 191, 192, 198
PSA, 197                                                   reciprocity, 199, 200, 203, 204
psoriasis, 8, 9                                            recognition, 53, 199, 219
psychiatric disorders, 32, 40, 41, 45, 56, 58, 102, 153    recommendations, xii, xix, 100, 130, 141, 142, 143,
psychiatry, 51, 244, 246                                       146, 181
psychoactive drug, xviii, 150                              recovery, 37, 216, 245
262                                                         Index
recreational, xvii, 10, 19, 20, 25, 33, 53, 65, 66, 88,      salicylates, 10
    90, 101, 102, 105, 109, 114, 130, 147, 148, 149,         saliva, 36, 68
    151, 175, 181, 184, 185, 193, 205, 207, 209, 223,        sample survey, 108
    224, 229                                                 sanctions, 221, 222
recruiting, 8, 9                                             scaling, 220
redistribution, 68                                           schizophrenia, 8, 34, 43, 44, 47, 48, 52, 58, 59, 179,
reflexes, 37                                                    181, 208, 223
reform, 33, 70, 223                                          schizotypy, 58
Registry, 186                                                school, xiv, 29, 37, 40, 45, 46, 59, 179, 181, 207,
regulations, xviii, xix, 90, 91, 103, 106, 111, 141,            209, 211, 212, 213, 214, 215, 216, 217, 218, 219,
    145, 183, 189, 200, 206, 222, 224                           241
rehabilitation, 220, 236, 239                                school performance, 181, 212
reinforcement, 57, 102, 197                                  school psychology, 219
relaxation, 142, 183                                         science, 12, 25, 82, 92, 145, 153, 186, 219, 230, 239,
relevance, 14, 73, 124, 128, 155, 174, 176                      245
relief, 7, 14, 23, 24, 49, 76, 85, 86, 97, 107, 110, 126,    scleroderma, 96
    129, 134, 135, 137, 138, 139, 141, 142, 143, 144,        sclerosis, 52, 135, 157, 162, 178, 222
    164, 170, 177, 178, 183, 200, 222, 228                   scope, 125, 131, 201
REM, 36                                                      second hand smoke, 203, 204, 205
remission, 8                                                 secretion, 208
renal failure, 191                                           security, 190, 199, 200
repressor, 22                                                sedative, 19, 20, 30, 66, 78
reproduction, 4                                              seedlings, 35, 53
reputation, 115, 117                                         seizure, 114
requirements, 23, 24, 140, 191, 195, 219                     selective serotonin reuptake inhibitor, 191
researchers, xviii, 31, 71, 72, 124, 141, 243                self-regulation, 121
resistance, 23, 38, 39, 77, 78                               self-report data, 29
resolution, 3                                                sellers, 35
resources, 4, 213, 223, 230                                  sensation, 106, 137, 218
respiratory problems, 10                                     sensation seeking, 218
response, 51, 74, 82, 91, 108, 127, 132, 143, 148,           sensations, 96
    160, 162, 163, 164, 165, 169, 193, 219                   sensitivity, 69, 137, 195, 198, 218
restrictions, 71, 203                                        sensor, 10
retail, 35                                                   sensory experience, 142
rheumatic fever, 11                                          serotonin, 67, 70, 106, 191
rheumatoid arthritis, 16, 34, 100, 103, 180, 195             serum, 192, 193
ribosomes, 77                                                services, xii, 50, 212, 213, 214, 216, 217
risk, 12, 14, 16, 23, 24, 27, 28, 31, 38, 39, 40, 41, 42,    sesquiterpenoid, 78
    43, 44, 45, 47, 51, 55, 58, 69, 93, 103, 120, 138,       sewage, 29, 50
    143, 147, 152, 179, 182, 183, 184, 185, 186, 191,        sex, 69, 208, 212, 218
    201, 204, 208, 211, 213, 215, 216, 218, 219, 223,        sexual abuse, 235
    230, 245                                                 sexual behavior, 218
RNA, 77                                                      sexual health, 37, 50
Romania, 30, 182                                             sexual intercourse, 97
Russia, 30                                                   sexuality, 244
                                                             shape, 181
                                                             short-term memory, 41, 78, 208
                           S                                 showing, 14, 124, 176
                                                             sickle cell, 195
safety, 5, 49, 52, 61, 91, 99, 102, 103, 109, 110, 123,
                                                             side chain, 72
   128, 153, 155, 156, 177, 182, 202, 205, 227, 228,
   230
                                                   Index                                                      263
side effects, 3, 11, 12, 14, 36, 51, 114, 120, 137, 143,   stroke, 33, 181
    155, 156, 176, 181, 192, 194, 196, 203, 209, 228       structure, 8, 9, 35, 55, 58, 70, 71, 79, 218, 229
signalling, 99                                             substance abuse, 41, 51, 93, 143, 186, 212, 216, 219
signs, 31, 215                                             substance use, 16, 40, 42, 56, 57, 59, 60, 91, 92, 183,
silk, 150                                                     212, 218, 219, 245
sinusitis, 36                                              substance use disorders, 16, 40, 56, 57, 60
skeletal muscle, 114                                       substitution, 23
skeleton, 80                                               substrates, 77
skin, 190                                                  suicidal ideation, 69
sleep deprivation, 96                                      suicide, 44, 45, 47, 58, 215, 223, 225, 235, 244
smoking, 10, 23, 27, 31, 35, 36, 38, 39, 40, 42, 44,       suicide rate, 223
    45, 46, 48, 51, 54, 55, 75, 76, 81, 88, 96, 103,       supervision, 45, 143
    109, 110, 121, 123, 129, 130, 131, 133, 137, 138,      suppliers, 33, 230
    140, 142, 147, 150, 152, 175, 176, 181, 194, 199,      supply chain, 33
    200, 201, 202, 203, 204, 208, 209, 211, 222, 231       suppository, 24
social anxiety, 34, 45, 59                                 suppression, 33, 35, 36, 114
social behavior, 47                                        survival, 4, 127
social change, 211                                         susceptibility, 41
social interaction, 67                                     Sweden, 30, 161, 241
social situations, 45                                      sweeteners, 23
society, 45, 217, 223, 229, 239                            swelling, 208
solubility, 24, 77                                         Switzerland, 30, 116
solvents, 150, 151                                         sympathy, 230
somnolence, 137, 139                                       symptoms, xix, 23, 39, 42, 44, 49, 53, 55, 57, 77, 85,
South America, 18                                             88, 89, 91, 92, 95, 96, 97, 99, 100, 122, 124, 127,
Southeast Asia, 51                                            140, 177, 179, 181, 195, 200, 201, 208, 209, 213,
Spain, 182                                                    218, 227, 228, 229, 245
spastic, 157                                               synaptic plasticity, 100
spasticity, 7, 26, 49, 52, 100, 103, 114, 135, 137,        synaptic transmission, 197
    138, 146, 148, 149, 151, 178, 180, 182, 201, 222       syndrome, 8, 9, 34, 37, 39, 42, 48, 49, 54, 55, 57, 58,
species, 4, 5, 7, 20, 53, 71, 74, 78, 147, 148, 149, 152      60, 96, 97, 102, 103, 114, 115, 123, 124, 126,
spinal cord, 67, 106, 145, 177                                127, 128, 148, 149, 181, 209, 210
spinal cord injury, 145, 177                               synergistic effect, 77
spleen, 67, 68                                             synthesis, 22, 79, 113, 125, 130
Spring, 100, 205                                           Synthetic cannabinoids, 35, 53, 66, 69, 70, 107, 108,
squamous cell, 38, 54                                         111, 131, 182, 192, 193, 196, 204
squamous cell carcinoma, 38, 54                            synthetic opioids, xvii
stability, 224
standardization, 109, 124, 125
starvation, 122
                                                                                     T
states, 11, 18, 28, 29, 105, 109, 145, 147, 148, 149,
                                                           T lymphocytes, 106
    179, 182, 183, 186, 190, 203, 207, 209, 223
                                                           tachycardia, 37, 181, 208
stigma, 99, 101, 102, 109, 202, 230
                                                           tar, 38, 110, 130
stigmatized, 103, 111, 202
                                                           target, 16, 32, 78, 122, 126, 213, 214, 215, 216
stimulation, 74, 91, 114, 116, 118, 120, 123, 125,
                                                           taxonomy, 102
    126, 127, 130, 148, 201
                                                           teachers, 215, 219
stimulus, 137
                                                           techniques, 12, 20, 23, 36, 45, 48, 66, 68
stomach, 19, 24, 74, 75
                                                           technologies, xviii
storage, 4, 35, 72, 79, 199, 202, 224
                                                           technology, 82
stress, 46, 48, 53, 78, 96, 100, 105, 106
                                                           teens, 184, 207
striatum, 32, 57
                                                           telephone, 45, 59, 149
264                                                        Index
ventilation, 206                                          weight loss, 97, 105, 110, 113, 114, 115, 116, 117,
Venus, xiv, 211                                              118, 119, 120, 122, 126, 127, 128, 182, 209
vertebrates, 99, 114                                      weight reduction, 115
vesicle, 74                                               weight status, 124
victims, 193, 235                                         welfare, 236, 239
violent behavior, 37, 57                                  well-being, 95, 97, 218, 236
violent crime, 218, 223                                   Western Europe, 19
viscosity, 23                                             whiplash, xix
volunteerism, 235                                         white matter, 41, 45, 218
vomiting, xiv, 7, 34, 37, 100, 105, 106, 107, 108,        withdrawal symptoms, 31, 35, 42, 43, 46, 57, 181,
   109, 110, 111, 114, 115, 126, 130, 148, 174, 181,         196, 209
   182, 209                                               workers, 202
vulnerability, 50, 227                                    working memory, 212
                                                          workplace, 202
                                                          World Anti-Doping Agency, 40
                         W                                World Bank, 16
                                                          World Health Organization (WHO), 4, 152, 193, 197
waiver, 202
                                                          worldwide, 11, 130, 207
walking, 97
war, 10
warts, 7                                                                            Y
Washington, 16, 25, 28, 51, 92, 219
waste water, 29                                           yield, 5, 20, 23, 122
water, 11, 24, 30, 37, 38, 54, 97, 124, 137, 150, 209     young adults, 16, 29, 36, 39, 42, 45, 47, 65, 128, 180,
weakness, 173, 181                                           185, 186, 201, 218
websites, 194                                             young people, 218
weight gain, 36, 120, 122, 123, 124                       Youth program, 216