Introduction To Tumor Virus
Introduction To Tumor Virus
● Virus Structure:
○ composed of genetic material (DNA/RNA) enclosed within a capsid protective (protein coat)
○ some possess an envelope made of phospholipids & proteins, derived from the host cell's membrane.
○ Capsid Shapes:
■ Icosahedral
■ Prolate (head-tail)
■ Filamentous
● 1. Viral Entry:
○ Virus surface proteins interact with host cell proteins for attachment/adsorption to the host cell
membrane.
○ A hole forms in the cell membrane → allows virus particles / genetic material to enter the host cell,
where replication can begin.
● 2. Viral Replication:
○ The virus takes control of the host cell's replication machinery.
○ distinction between susceptibility & permeability is made
○ Permissibility of the host cell determines the infection's outcome.
○ Once control is established, the virus replicates rapidly, producing millions of copies.
● 3. Viral Shedding: (final stage)
○ After exhausting the host cell's resources, the cell often dies.
○ Newly produced viruses are released (shedding) to find new hosts.
● 4. Viral Latency (Lysogenic):
○ Some viruses can "hide" within a cell to evade host defenses / continuous replication isn't beneficial.
○ virus is inactive & produces no progeny until an external stimulus (light, stress) prompts activation.
● Structure of RSV:
○ env gene: Encodes glycoprotein spikes for virion adsorption to the cell surface.
○ gag gene: Encodes core proteins forming the virus's protein shell.
○ pol gene: Encodes reverse transcriptase & integrase.
○ Genome: Two identical copies of genomic RNA.
● RSV-induced Focus:
○ Infected cells become rounded, overgrow, pile up, forming a "focus."
○ Surrounding uninfected cells exhibit contact inhibition & stop proliferating.
● Transformed Cells by Virus Forming Foci:
○ Normal fibroblast cells display contact inhibition (density inhibition).
○ Transformed cells lose this inhibition and grow on top of each other.
○ a group of descendant cells is a "cell clone," and the focus is a "clonal growth."
○ Cell transformation by RSV resembles tumor appearance ; the program is activated upon infection.
● Temperature-sensitive Mutant (Berkeley Experiment):
○ Mutant viruses lose function at high temperatures (denaturing mutant proteins).
○ Viral genome copies persist in cells (replicating with cell proliferation).
○ The continued action of the virus is required for both the initiation & maintenance of the
transformed phenotype.
SV40 Virus
● Characteristics:
○ Icosahedral symmetry.
○ dsDNA genome.
○ Discovered in 1953 (monkey virus).
○ Causes cytopathic effect: vacuoles (in permissive cells).
○ Nonpermissive in rodent hosts (doesn't complete lytic cycle but can transform cells).
○ Able to seed tumors in vivo (in nonpermissive hosts).
● SV40 Genome (circular dsDNA):
○ Form I (kinked): Closed, circular, super-coiled dsDNA.
○ Form II (open spread circle): Relaxed, nicked circular dsDNA.
○ Form III: Linear form.
● Property of Transformed Cells:
○ Anchorage Independence: Transformed cells can form spherical colonies in soft agar gel, unlike
normal cells which require attachment to a solid substrate to grow.
● Test of Tumorigenicity:
○ To check if cells are fully transformed & have neoplastic traits:
■ Inject in vitro transformed cells into a syngeneic host mouse.
■ Inject into an immunocompromised host mouse (nude mouse).
● Expression of the SV40 T antigen:
○ The large T antigen (LT) is expressed in the nuclei of transformed cells.
○ BK virus (an SV40 relative) infection caused transformation & tumorigenesis in the bladder of a
kidney transplant patient.
● How is the viral genome replicated/maintained during transformation?
○ If the viral genome were lost during cell proliferation, transformation might not be maintained
○ Viral genomes typically lack centromeres for proper allocation during cell division.
○ In a lytic cycle, the viral genome exists as extrachromosomal molecules, replicating independently
of the host genome, producing many copies.
● SV40 Genome is Integrated into Host Genome (in transformed cells):
○ SV40 DNA from free virus sediments more slowly (lower molecular weight) than SV40 DNA from
transformed cells.
○ In transformed cells, SV40 DNA is covalently associated with the high molecular weight
chromosomal DNA of the host cells.
○ Formation of Integrated SV40 Genome:
■ Occurs with low efficiency.
■ Involves non homologous (illegitimate) recombination.
■ Integration occurs at random sites in the host genome.
■ Results in a covalent linkage of viral and host genomic DNA.
Oncogenes and Proto-oncogenes
Drug specific to 1 drug / narrow class of drugs broad range of structurally & mechanistically
spectrum unrelated drugs
Primary cause alterations in drug target / pathway general mechanisms like increased drug efflux,
altered metabolism, enhanced DNA repair,
reduced apoptosis
Implication may still respond to other drugs with different limits treatment options ; high failure rate
mechanisms
● Decreased Uptake
○ Mediated by SLC transporters (>300 membrane-bound proteins)
○ SLCs transport diverse substrates (nutrients, drugs, xenobiotics) ; About 40 SLC genes are involved
in uptaking specific cancer drugs.
○ Mutations in SLCs can affect drug efficacy by:
■ Causing misfolded proteins leading to degradation.
■ Preventing trafficking of the transporter to the cell surface.
■ intracellular accumulation of misfolded protein, triggering the unfolded protein response.
● Increased Efflux
○ Mediated by ABC transporters (superfamily of 48 transporters).
○ transporters use energy from ATP hydrolysis to move substrates across membranes (uptake/export)
○ P-glycoprotein (P-gp) / MDR1 / ABCB1 / CD243:
■ An important ATP-dependent efflux pump with broad substrate specificity, pumping many
foreign substances out of cells.
■ Evolved as a defense mechanism; found in animals, fungi, and bacteria.
■ Expressed in liver, pancreas, kidney, colon, jejunum, and brain capillary endothelial cells.
■ Role in Multi-drug Resistance: P-gp and other ABC transporters extrude numerous chemical
compounds, leading to:
■ Failure of cancer chemotherapy (MDR).
■ Prevention of carcinogen accumulation in healthy tissues.
■ Affecting ADME of drugs.
■ Protection from environmental toxins.
■ Acquired Resistance: Meta-analysis showed MDR1 mRNA and protein levels increase
significantly after chemotherapy, indicating a role in acquired resistance.
■ Intrinsic Resistance: Overexpression is frequent in tumors with inherent resistance. MDR1
gene amplification is rare in clinical samples.
■ P-gp in Clinical Carcinogenesis: Lung carcinomas in smokers often express more P-gp and
are more drug-resistant.
■ Clinical Significance: ~50% of human cancers express P-gp at levels sufficient for MDR.
P-gp inhibitors have shown limited success.
■ P-gp is sufficient for MDR but may not be necessary or the sole cause.
● Reduced Apoptosis: Cancer cells become less sensitive to drug-induced cell death.
● Altered Cell Cycle Checkpoints: Changes allow cells to bypass drug-induced arrest.
● Altered Growth Pathways: Activation of alternative survival pathways.
● Increased Metabolism of Drugs: Drugs are inactivated more rapidly.
● Increased or Altered Targets: More target molecules need to be inhibited, or the target is mutated.
● Increased Repair of Damage: Cells become more efficient at repairing drug-induced damage.
● Compartmentalization: Drugs are sequestered away from their targets.
Mutations Associated with Drug Target
● Dysregulated Apoptosis: Reduced efficiency of linking drug-induced DNA damage to cell death.
● DNA Damage Repair:
○ Many chemotherapies induce DNA damage (platinum drugs, topoisomerase inhibitors).
○ Increased DNA damage repair capacity in cancer cells leads to resistance.
○ MMR: Mutations in MMR genes (MLH1, MSH2) can cause resistance to various cytotoxic
chemotherapies.
○ Genomic Instability: A hallmark of cancer, contributing to heterogeneity and resistance.
○ PARP Inhibitors: Target the DNA repair enzyme PARP ; show synthetic lethality in tumors with
BRCA1/BRCA2 mutations (impaired homologous recombination).
■ Resistance to PARP Inhibitors: occur due to in-frame deletions in BRCA2 that partially
restore its DNA repair function.
● Activation of Prosurvival Adaptive Responses:
○ Cytotoxic drugs can activate prosurvival signals, morphological changes (EMT), autophagy.
Causes of Cancer
● Age, Chemical Exposure, Diet, Radiation, Infection, chronic inflammation, obesity, smoking
● Genetic Factors
○ Oncogenes and Tumor Suppressors:
■ Proto-oncogenes: Normal genes that can become oncogenes when mutated (MYC, ErbB2,
PI3KCA, CCND1)
■ TSG: Normally prevent cancer by inhibiting cell division / causing cell death. Mutations
lead to loss of this protective function. (P53, BRCA1/2, PTEN)
○ Non-coding Mutations: The majority of somatic variants in cancer genomes. Mechanisms include
changes in transcription factor binding, miRNA binding sites, and higher-order chromatin structure
○ Epigenetic Mutations: Changes in histone & DNA modifications
■ Driver Mutations in Histone Modifications: Histone H3.3 mutations in gliomagenesis.
■ G34R/V: More prevalent in cerebral hemispheres, older patients; inhibits SETD2
function (H3K36me3).
■ H3.3K27M: Primarily in midline locations, younger patients; inhibits EZH2
function (H3K27me3).
Conventional Cancer Therapy
Surgery Therapy
Radiotherapy
● Uses high-energy particles/waves (X-rays, gamma rays, electron beams, protons) to destroy/damage cancer
cells by making small breaks in their DNA, preventing growth/division and causing death.
● Goals of Radiotherapy
○ Cure or shrink early-stage cancer.
○ Prevent recurrence.
○ Treat symptoms of advanced cancer (palliative).
○ Treat recurrent cancer (re-assessment needed for same area).
● Ways Radiotherapy is Given
○ External Radiation (External Beam): Machine directs rays from outside the body to the tumor.
Usually given over many weeks as outpatient visits.
○ Internal Radiation (Brachytherapy): Radioactive source placed inside body, in or near the tumor.
○ Systemic Radiation: Radioactive drugs given orally / intravenously, traveling throughout the body.
● Issues of Ionizing Radiation
○ Early Side Effects: Fatigue, skin problems, hair loss, nausea.
○ Late Side Effects: Scar tissue affecting lung/heart function; bladder, bowel, fertility, sexual
problems (if pelvis/belly treated); can cause a second cancer.
○ Kills all fast-growing cells, including some normal ones.
Chemotherapy
● Uses powerful drugs to shrink or kill cancer cells. Works throughout the whole body, so can kill
metastasized cancer cells far from the primary tumor
● Main Goals
○ Cure.
○ Control.
○ Ease symptoms (Palliation).
● Determining Which Drugs to Use
○ Type and stage of cancer.
○ Patient's age and overall health.
○ Other health problems
○ Past cancer treatments.
● Approaches to Treatment (Delivery Methods)
○ Intravenous (IV): Most common, infused into a vein.
○ Oral: Pills
○ Topical: Cream/lotion for skin cancer.
○ Direct Placement: Injected into spinal fluid or brain (via a device).
○ HIPEC: For cancers spread to abdominal cavity lining (appendix, colon, stomach, ovaries).
Delivered directly into the abdomen after surgery in the OR
■ Prevents remaining cells from growing into new tumors. Higher chemo concentration
delivered more effectively and safely than standard IV chemo for abdominal cancers. Better
at killing microscopic cancer cells. Fewer side effects due to the peritoneal plasma barrier
● Different Types of Chemotherapy
○ Hormone Therapies: Prevent hormone production or interfere with hormone action (e.g.,
Tamoxifen for ER+ breast cancer).
○ Signal Transduction Inhibitors: Target continuous cell division signals (e.g., Trastuzumab for
HER2+ gastric cancer).
○ Gene Expression Modulators: Modulate genes controlling cell cycle/apoptosis (e.g., Romidepsin,
a histone deacetylase inhibitor).
○ Apoptosis Inducers: Cause cancer cell death by bypassing their strategies to avoid apoptosis.
○ Angiogenesis Inhibitors: Block tumor growth by interfering with the formation of new blood
vessels needed for oxygen and nutrients.
○ Monoclonal Antibodies that Deliver Toxic Molecules: Antibody binds to target cell, delivering a
linked toxin (radioactive substance, chemical) to kill the cell.
● Risks of Chemotherapy
○ Chemo drugs kill fast-growing cells, affecting normal fast-growing cells too.
○ Side Effects: Fatigue, hair loss, easy bruising/bleeding, infection, anemia, nausea/vomiting, appetite
changes, fertility problems.
○ Some chemo/radiation works by producing free radicals. Antioxidant vitamins (A, E, C) can prevent
free radical formation, so should not be taken unless advised by a doctor.
Hormone Therapy
● Systemic therapy that adds, blocks, or removes hormones to slow/stop cancer cell growth
● Usually involves medications preventing cancer cells from getting hormones needed for growth.
● Treatment option for prostate, breast, ovarian, uterine, or thyroid cancers.
● Factors influencing choice: age, tumor type/size, presence of hormone receptors on the tumor.
● Hormone Therapy for Breast Cancer
○ Used if tumor growth depends on Estrogen or Progesterone (ER/PR positive).
○ Tamoxifen: An ER blocker, prevents estrogen uptake by cancer cells.
● Side Effects of Hormone Therapy
○ Breast Cancer Patients: Hot flashes, vaginal issues, decreased sex drive, mood changes.
○ Prostate Cancer Patients: Reduced erectile/orgasmic ability.
○ Aromatase Inhibitors: Joint/muscle pain, increased risk of osteoporosis.
Targeted Therapy (2nd Generation)
● Definition: Uses a patient’s own T lymphocytes with anti-tumor activity, expanded in vitro, and reinfused
into the patient
● Highly effective for metastatic melanoma Types of ACT: TILs, TCRs, CARs
TILs
● Source: Immune cells found in tumor biopsies, conditioned to the tumor microenvironment. Abundance
varies by tumor type/stage and can relate to prognosis
● Process
○ Collect autologous TILs from patient tumors.
○ Detect lymphocytes (e.g., using CD3 marker or gene expression methods like Microarray/RNA
Sequencing with deconvolution).
○ Activate and expand T cells using cytokines (e.g., IL-2).
○ Select TIL lines with best tumor reactivity for further expansion in a REP using anti-CD3 activation.
○ Optional: Preliminary chemotherapy regimen (7 days before TIL infusion) to deplete endogenous
lymphocytes, giving transferred TILs better access to tumor sites.
● Clinical Success
○ Induces complete, durable regression of metastatic melanoma
○ In colorectal cancer, associated with microsatellite instability cancers and effective immune
checkpoint inhibitor therapy in GI cancers.
○ Associated with better outcomes in epithelial ovarian cancer.
○ Combination with prior immunotherapy (e.g., IL-2, anti-CTLA4) shows higher, more durable
responses, suggesting synergy.
TCR Therapy
● Rationale: For patients whose T cells haven't recognized their tumors or whose T cells can't be sufficiently
activated/expanded . Allows for personalized treatment by choosing optimal targets and T cell types.
● T-cell Receptor (TCR) Structure and Function
○ Antigen receptors on T cells.
○ Disulfide-linked membrane-bound heterodimer (usually α and β chains, or γ and δ chains)
complexed with invariant CD3 chains (CD3δ, CD3ε, CD3γ).
○ Two T cell subsets: γδ TCR (minor), αβ TCR (major).
○ Antigenic diversity from random rearrangement of V, D, J gene segments. TCR α chain: V, J, C
segments. TCR β chain: V, D, J, C segments.
○ Recognizes peptide fragments of antigen bound to MHC molecules.
● Activation of T cells: Requires TCR binding to MHC-presented antigen AND CD28 (on T cell) binding to
B7 (CD80/86 on antigen-presenting cell)
● Development of Engineered TCR-T cells
○ Choose Tumor Antigen: TAA or tumor-specific endogenous protein presented by tumor cell MHC.
○ High Affinity T cell Clone Isolation: Isolate high affinity T cells clone for the chosen tumor
antigen.
○ TCR α/β Gene Cloning & Packaging: Clone the TCR α/β genes from these high-affinity T cells.
Use retro- or lenti-viruses to deliver these genes into donor T cells from the patient's peripheral
blood.
○ Improving TCR Performance:
■ Enhance TCR affinity (e.g., amino acid substitutions in CDRs).
■ Increase TCR expression (e.g., murine-human hybrid TCR).
■ Increase stability of CD3ζ/TCR complex.
■ Reduce mispairing of TCR chains (e.g., add a second disulfide bond between α and β
constant regions, add cysteines, use mouse TCR genes).
■ Activation with cytokines like IL-2.
○ Adoptive Transfer: Infuse engineered T cells back into the patient.
● Advantages and Disadvantages of TCR-T
○ Advantages: Function through well-understood T cell signaling pathways; natural way the body
fights foreign elements.
○ Disadvantages: Restricted to one HLA type; α/β TCRs cannot target non-protein TAAs (e.g.,
carbohydrate or lipid antigens).
CAR-T
● Rationale: Overcomes the limitation of TIL and TCR-T therapies which require antigen presentation by
MHC
● Patient’s T cells are equipped with a synthetic receptor (CAR).
● Workflow of CAR-T: (Diagrammatic representation, general steps include T cell collection, engineering,
expansion, infusion)
● Engineered CAR Structure
○ Antigen Recognition Targeting Moiety: Typically a scFv derived from an antibody.
○ Hinge/Spacer: Connects scFv to the transmembrane domain.
○ Transmembrane (TM) Domain: Anchors CAR in the T cell membrane.
○ Signaling Domains: Commonly CD3ζ (primary activation signal).
○ Co-stimulatory Domains: (e.g., CD28, 4-1BB) enhance T cell activation, proliferation, persistence.
● Alternative CAR-T Strategy (Universal)
○ Uses CAR T-cells expressing a high-affinity receptor for a molecule like FITC.
○ A two-part CAR-T adaptor molecule (CAM) is used: FITC conjugated to a tumor-homing ligand.
This allows the CAR-T cell to be directed by the adaptor.
● Advantages of CAR-T Therapy
○ Can bind to cancer cells even antigens aren't presented via MHC (unlike TCR-T which requires
APCs).
○ However, CAR T cells can only recognize antigens naturally expressed on the cell surface, so the
range of potential targets is smaller than with TCRs.
○ Being explored for many cancer types. "Off-the-shelf" CAR-T cells from stem cells are in clinical
trials for timelier treatment.
TILs TCR-T CAR-T
source of T Harvested directly from the Typically T cells isolated from Typically T cells isolated from
cells patient's tumor biopsies; these the patient's peripheral blood the patient's peripheral blood
cells are already "conditioned via leukapheresis
to the tumour
microenvironment"
mechanism Relies on the T cells' naturally Uses T cells genetically Uses T cells genetically
of tumor occurring TCRs that have engineered to express a new engineered to express a
recognition already recognized antigens TCR that has high affinity for synthetic CAR. The CAR's
within the tumor a specific tumor antigen antigen-binding domain
microenvironment. presented by MHC molecules (usually an scFv from an
antibody) directly recognizes
antigens on the cancer cell
surface
genetic Primarily involves selection Requires genetic engineering Requires genetic engineering
engineering and expansion of naturally to introduce the genes for the to introduce the gene encoding
tumor-reactive T cells. new, specific TCR α/β chains the synthetic CAR
Target Peptides derived from tumor Peptides derived from Primarily surface antigens.
antigen type antigens presented on MHC intracellular or surface tumor Cannot target IC antigens
molecules antigens presented on MHC unless a fragment is expressed
molecules.. Cannot target on the surface. The range of
non-protein antigens like potential antigen targets is
carbohydrates or lipids described as smaller than with
TCRs because they must be on
the cell surface
Need for Relies on T cells that have Can be used even if the the recognition specificity is
Pre-existing already infiltrated and patient's T cells have not engineered, so pre-existing
Tumor potentially recognized the naturally recognized their recognition by the patient's T
Recognition tumor. tumors, as the recognition cells is not required for the
by Patient's T specificity is engineered CAR to function.
cells
Key Effective for metastatic Can be personalized by Ability to bind cancer cells
advantage melanoma, can induce choosing optimal targets; even if antigens aren't
complete and durable functions through natural T presented via MHC,
regression cell signaling pathways potentially making more
cancer cells vulnerable
Key Process can be lengthy; not all Restricted to one HLA type; Can only recognize antigens
limitation patients have sufficient TILs cannot target non-protein naturally expressed on the cell
or TILs that can be effectively antigens surface; range of potential
expanded. targets smaller than TCRs in
terms of intracellular origin
Side Effects of Adoptive Cell Therapy
Cytokine Treatment
● Mechanism: Designed to alter immune homeostasis within tumor & provoke/ disinhibit immune effectors
● FDA Approved Examples
○ IFN-α: For metastatic renal cell carcinoma and melanoma. Works to activate NK cells.
○ IL-2: For metastatic renal cell carcinoma and melanoma. Is a T cell growth factor.
Immune Checkpoint Therapy
● Immune Checkpoints: Regulators of the immune system crucial for self-tolerance, preventing
indiscriminate attacks on normal cells. They need to be activated (or inactivated) to start an immune
response
● Cancer cells can use these checkpoints to avoid immune attack. When checkpoint proteins bind their
partners, they send an "off" signal to T cells.
● The immune system uses checkpoints to distinguish normal ("self") from "foreign" cells.
● Mechanism of Immune Checkpoint Inhibitors: Block checkpoint proteins from binding their partners,
preventing the "off" signal and allowing T cells to kill cancer cells. These inhibitors don't act directly on the
tumor but "take the brakes off" an existing immune response
● Known Checkpoint Proteins : CTLA-4 (CD152), PD-1 (CD279), LAG-3 (CD223), TIM-3 (HAVcr2),
KIRs (CD158), 4-1BB (CD137), GITR (CD357).
● Mechanism
○ T cell activation requires TCR engagement with antigen + CD28 (on T cell) co-stimulation by
binding CD80 (B7-1) / CD86 (B7-2) on APCs.
○ CTLA-4 is a homolog of CD28.
○ CTLA-4 on T cells binds B7 proteins with ~20 times greater affinity than CD28, outcompeting
CD28 (Inhibition and Ligand competition).
○ Excess soluble CTLA-4 can bind B7 on APCs, restricting B7 availability for CD28 binding
(Unresponsiveness).
● CTLA-4 Inhibitor
○ Blocking CTLA-4 has therapeutic benefits.
○ Ipilimumab: FDA approved for melanoma (2011).
○ Tremelimumab: (Not yet approved).
● PD-L1 and PD-L2 (bind to PD-1): Found on tumor cells and APCs.
● B7-1 and B7-2 (bind to CTLA-4): Found on APCs.
Side Effects of Immune Checkpoint Inhibitor Therapy
TCV