TOFA STATs
TOFA STATs
Madeline E. Kavanagh1,2, Benjamin D. Horning1,3, Roli Khattri3, Nilotpal Roy3, Justine P. Lu3,
Landon R. Whitby3, Jaclyn C. Brannon3, Albert Parker3, Joel M. Chick3, Christie L. Eissler3,
Ashley Wong3, Joe L. Rodriguez3, Socorro Rodiles3, Kim Masuda2, John R. Teijaro4, Gabriel M.
Simon3, Matthew P. Patricelli3*, Benjamin F. Cravatt2*
1
These authors contributed equally, 2Department of Chemistry, Scripps Research, La Jolla, CA
92037, USA; 3Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA 92121, USA;
4
Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla,
1
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
Abstract
The JAK family of non-receptor tyrosine kinases includes four subtypes (JAK1, JAK2, JAK3,
and TYK2) and is responsible for signal transduction downstream of diverse cytokine receptors.
JAK inhibitors have emerged as important therapies for immuno(onc)ological disorders, but their
use is limited by undesirable side effects presumed to arise from poor subtype selectivity, a
common challenge for inhibitors targeting the ATP-binding pocket of kinases. Here, we describe
the chemical proteomic discovery of a druggable allosteric cysteine present in the non-catalytic
pseudokinase domain of JAK1 (C817) and TYK2 (C838), but absent from JAK2 or JAK3.
transphosphorylation and cytokine signaling, while appearing to act largely as “silent” ligands for
TYK2. Importantly, the allosteric JAK1 inhibitors do not impair JAK2-dependent cytokine
signaling and are inactive in cells expressing a C817A JAK1 mutant. Our findings thus reveal an
2
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
neurodegenerative diseases, and cancer1,2. More than 50 different cytokines signal through a
family of non-receptor Janus tyrosine kinases (JAKs), which, in humans, consists of JAK1,
JAK2, JAK3, and TYK21,3. JAKs associate with the intracellular tail of specific cytokine receptors
receptor, and downstream signaling proteins, including the STAT family of transcription factors.
The specific combination of JAK enzymes and STAT transcription factors that are activated by a
given cytokine is cell-type and context-dependent, allowing the JAK-STAT system to regulate
The key role of JAK-STAT pathways in immunology and cancer has motivated the
pursuit of JAK inhibitors, and many pan-JAK inhibitors have been described1,4. These
compounds have provided preclinical and clinical evidence that inhibiting JAK-STAT signaling
can alleviate aberrant cytokine responses and have established JAKs as important therapeutic
targets1,4. There are currently seven FDA-approved JAK inhibitors for the treatment of
diseases (e.g. polycythemia, leukemia and GVHD), and one compound (baracitinib) that has
emergency use authorization (EUA) for COVID-194. All FDA-approved JAK inhibitors act by an
orthosteric mechanism, meaning that they bind to the conserved ATP pocket of the kinase
domain, and, even though individual compounds have differing relative selectivity profiles
across the JAK family, they all inhibit more than one JAK isoform with moderate-to-high potency
(IC50 < 1 μM)5–12. This lack of selectivity has important translational implications, as there is
growing concern over an array of adverse side effects caused by JAK inhibitors1,3,4,13, including
3
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
elevated liver enzymes3. In 2021, these concerns prompted the FDA to place a “black box”
warning on JAK inhibitors indicated for chronic conditions such as rheumatoid arthritis15.
The lack of subtype selectivity of FDA-approved JAK inhibitors may contribute to their
adverse clinical effects16,17, and there is accordingly considerable interest in the discovery of
JAK inhibitors with improved specificity. Such compounds might not only constitute next-
generation therapeutics, but would also serve as valuable research tools to better understand
the unique contributions made by each JAK isoform to physiology and disease. Subtype-
selective JAK inhibitors have been pursued by multiple strategies. For example, covalent
inhibitors of JAK3 have been developed, such as ritlecitinib, that target a cysteine (C909)
uniquely found in the activation loop of this kinase compared to other JAKs18,19. While this
approach achieves specificity for JAK3 over other JAKs, ritlecitinib cross-reacts with TEC family
kinases, which also share a cysteine at an equivalent position. JAKs are distinguished from
many other kinases by having an additional non-catalytic pseudokinase (JH2) domain that
regulates kinase activity and is a hotspot for gain- or loss-of-function mutations2,20. Notably,
compounds binding to the ATP pocket of the JH2 domain of TYK2 have been found to inhibit
this kinase with remarkable functional selectivity over JAK1-JAK321,22, and one of these agents,
disorders21,23,24.
selective JAK1 inhibitors are still lacking. Although some orthosteric JAK1 inhibitors have been
reported that display improved subtype selectivity, these compounds (e.g., abrocitinib, filgotinib)
still generally show substantial cross-reactivity with JAK2 (e.g., < 1 µM IC50 values), depending
inhibitors of JAK1 is an important objective, as several lines of evidence indicate that JAK1
blockade contributes to the efficacy of pan-JAK inhibitors in chronic autoimmune disorders. For
4
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
instance, gain-of-function JAK1 mutations promote multi-organ immune dysregulation27 and are
associated with specific types of cancer (e.g., leukemia28,29 and gynecological tumors30), while
broadly expressed and plays essential and non-redundant roles downstream of class II, γc, and
gp130 cytokines32, many of which are dysregulated in inflammatory diseases2. Nonetheless, the
precise contribution of JAK1 to homeostatic immune function and disease remains only partly
understood due to a lack of genetic models and selective chemical tools. JAK1 deletion is
perinatal lethal to mice32, and consequently, much of our understanding of JAK1 biology has
relied on studies with conditional knockout mice lacking JAK1 in specific cell types33–35, JAK1-
the pseudokinase domain of JAK1 (C817) and TYK2 (C838) but absent from JAK2 and JAK3.
TYK2_838 with high potency and proteome-wide selectivity and show that this agent blocks
JAK1 signaling in human cancer cell lines and primary immune cells, while sparing JAK2-
dependent pathways. VVD-118313 does not inhibit signaling of a C817A JAK1 mutant and
appears to act as a silent ligand for TYK2 in the context of primary immune cells. Mechanistic
studies indicate that VVD-118313 does not inhibit the catalytic activity of purified JAK1, but
potently blocks JAK1 trans-phosphorylation in cells. Integrating our findings with previous work
on allosteric TYK2 inhibitors, such as BMS-986165, points to the potential for leveraging
multiple druggable pockets in the pseudokinase domain of JAKs to develop inhibitors with
5
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
Results
ligandable cysteine shared by JAK1 (C817) and TYK2 (C838)41. Both cysteines were
Data Fig. 1a)41, as determined by mass spectrometry (MS)-ABPP experiments that monitored
cysteines in the human T-cell proteome (Fig. 1a). Other quantified JAK1 and TYK2 cysteines
were unaffected in their IA-DTB reactivity by KB02 or KB05 treatment (Fig. 1b and Extended
JAK1_C817 and TYK2_C838 are located in the catalytically inactive pseudokinase (JH2)
domain shared across the JAK family (Fig. 1c). The JH2 domain has been found to regulate the
kinase activity of the JH1 domain through allosteric mechanisms and is a hotspot for gain- or
loss-of-function mutations (Fig. 1c)2,20,42–44. We noted that other JAK family members – JAK2
and JAK3 – did not share the ligandable cysteine (Fig. 1d). A closer examination of the JAK1
JH2 crystal structure in comparison to other kinase structures revealed that C817 is in the C-
lobe proximal to a pocket formed by helices aE-F and aH-I, which, in the structurally related
kinase ABL, binds an auto-inhibitory N-terminal lipid (myristoylation) modification45–47 (Fig. 1e).
This pocket in ABL is targeted by the allosteric inhibitor asciminib, which stabilizes the inactive
conformation of the kinase48 and has recently been approved for the treatment of chronic
myeloid leukemia49. Even though asciminib is a reversible inhibitor, and JAK1 and TYK2 are not
required for myristoylation50), the proximity of C817/C838 to a pocket that has been exploited to
6
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
create allosteric drugs of another kinase encouraged us to further characterize the potential
We pursued the discovery of more potent and selective covalent ligands for
fragment hit 1a (Fig. 1f) that showed target engagement values (TE50s) of 2.1 µM and 45 µM for
JAK1_C817 and TYK2_C838, respectively (Fig. 1g and Table 1). Using a homogeneous time-
resolved fluorescence (HTRF) assay in human peripheral blood mononuclear cells (PBMCs),
dependent cytokine pathway – with an IC50 value of 1.4 µM (Fig. 1h and Table 1), suggesting
that covalent ligands targeting JAK1_C817/TYK2_C838 acted as JAK1 and/or TYK2 inhibitors.
The corresponding racemate of 1a (compound 1) was ~two-fold less active in both HTRF and
based on the greater potency displayed by 1a for JAK1_C817 over TYK2_C838, screened
these compounds for: i) in vitro engagement of JAK1_C817 (TE50) in human cell proteomes by
targeted MS-ABPP; and ii) cell-based functional activity (IC50) on JAK1-dependent signaling
compound interactions with JAK1_C817 by three orders of magnitude and observed a strong
correlation (R2 ~ 0.93-98) between these in vitro TE50 values and the in situ IC50 values for
blocking STAT phosphorylation (Fig. 2b, c and Table 1), further supporting that the compounds
potency included the addition of a second chlorine atom at the meta-position of the phenyl ring
7
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
(e.g., compound 3) and replacement of the terminal methyl of the butynamide with a pyrrolidine-
methylsulfonamide (e.g., compound 5). The tested compounds generally showed >10-fold
greater potency for engagement of JAK1_C817 compared to TYK2_C838 (Table 1). Separation
of the stereoisomers of compound 5 revealed that the enantiomers(S, R)-5a and (R, S)-5b were
substantially more potent than the corresponding diastereomers (5c and 5d), blocking STAT
phosphorylation with IC50 values (~0.03-0.05 µM) that were superior to the pan-JAK inhibitor
tofacitinib (Fig. 2c and Table 1). Compound 5a, hereafter referred to as VVD-118313, was
selected for further functional characterization as the compound showed the strongest overall
human PBMCs. Across >14,000 quantified cysteines, JAK1_C817 was the most potently
engaged site by VVD-118313 (0.01 – 10 µM, 3h), followed by TYK2_C838, with both cysteines
showing near-complete blockade in their IA-DTB reactivity in cells treated with 0.1 µM of VVD-
118313 (Fig. 2d, e and Supplementary Dataset 1). Two additional cysteines (HMOX2_C282,
(1 µM; Fig. 2d, e). Similar results were obtained in MS-ABPP experiments that analyzed the in
vitro proteome-wide reactivity of VVD-118313 in PBMC lysates, where JAK1_C817 was again
the most potently engaged cysteine, followed by TOR4A_C21, a site that was also engaged in
situ, albeit more weakly, and TYK2_C838 (Extended Data Fig. 2 and Supplementary Dataset
1). Taken together, these chemical proteomic data support that VVD-118313 is a highly potent
recombinantly expressed WT-JAK1 and a C817A-JAK1 mutant in the 22Rv1 human prostate
8
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
cancer cell line, which has a frameshift mutation in the JAK1 gene and consequently only
expresses JAK2 and TYK2 (JAK3 is immune cell-restricted in its expression)3. We also
evaluated a C810A-JAK1 mutant, as the quantified C817 tryptic peptide in our MS-ABPP
experiments also contained C810, a residue that is not conserved in TYK2 (Fig. 1d) and is
further away than C817 from the pocket predicted to bind electrophilic compounds (Fig. 1e). We
first treated 22Rv1 cells expressing the JAK1 variants with an alkynylated analogue of VVD-
118313 (alkyne probe 6 (0.1 µM, 2 h); Fig. 2f) and, after cell lysis, detected 6-labeled proteins
group, followed by SDS-PAGE and in-gel fluorescence scanning. Alkyne probe 6 reacted with
WT- and C810A-JAK1, but not C817A-JAK1, and the labeling of WT-JAK1 was blocked in a
heterodimerizing with other JAK family members (Fig. 3a)3. We selected a representative
subset of these pathways (IFNα-STAT1 and IL-6-STAT3), along with a JAK2-mediated pathway
recombinantly expressed WT and mutant forms of JAK1 in 22Rv1 cells. We first verified that
recombinant WT-JAK1 and the C810A and C817A mutants equivalently rectified intrinsic
defects in IFNα and IL-6 signaling in parental 22Rv1 cells37,52, as reflected by the greater IFNα
to mock cells (Fig. 3b and Extended Data Fig. 3a). We also noted that all of the JAK1 variant-
expressing cells displayed a similar degree of constitutive phosphorylation of the JAK1 JH1
activation loop (Y1034/Y1035) that was not further increased by cytokine treatment (Fig. 3b).
9
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
expressing cells (Fig. 3b, c). In contrast, the orthosteric JAK inhibitor tofacitinib equivalently
JAK1 (Fig. 3b, c). Interestingly, VVD-118313 also completely blocked the constitutive
phosphorylation of WT- and C810A-JAK1 but did not affect the phosphorylation of C817A-
JAK1(Fig. 3b, d). In contrast, tofacitinib only partly (~50%) reduced phosphorylation of all JAK1
variants (Fig. 3b, d). VVD-118313 and tofacitinib further differed in their effects on JAK2-
mediated signaling, where VVD-118313 was inactive, while tofacitinib fully inhibited PRL-
induced STAT5 phosphorylation (Fig. 3b, c). Concentration-dependent analyses revealed that
VVD-118313 maximally inhibited IFNα-STAT1 and IL-6-STAT3 phosphorylation (> 80% in each
case) in WT- or C810A-JAK1-expressing 22Rv1 cells at ~0.2 µM, while showing negligible
impact (< 10%) in cells expressing C817A-JAK1 up to 2 µM (Fig. 3e and Extended Data Fig.
3b, c). VVD-118313 inhibited WT- and C810A-JAK1 phosphorylation with even greater potency
than STAT1/STAT3 phosphorylation, showing maximal activity (> 90% blockade) at 0.05 µM
(Fig. 3e), which could indicate that only a small fraction of residually phosphorylated and
22Rv1 cells. Together, these data indicate that VVD-118313 acts as a selective allosteric
alkyne probe 6 reacted with recombinantly expressed WT-TYK2, but not a C838A-TYK2 mutant,
in 22Rv1 cells, and that pre-treatment with VVD-118313 blocked compound 6 reactivity with WT
TYK2 (Extended Data Fig. 4a). We did not observe a signal for endogenous TYK2 in these
experiments (e.g., in mock 22Rv1 cells), indicating that the expression level of TYK2 in 22Rv1
cells was too low to visualize with alkyne probe 6. 22Rv1 cells expressing recombinant WT- or
mock 22Rv1 cells (Extended Data Fig. 4b), suggesting that atypical TYK2 homodimers and/or
10
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
TYK2-JAK2 heterodimers can partly support IFNα signaling in these cells37,52,53. VVD-118313
but not C838A-TYK2, while the TYK2 inhibitor BMS-986165 blocked IFNα-STAT1
phosphorylation in both cell populations (Fig. 3f). We further noted that VVD-118313 and BMS-
986165 blocked the weaker IFNα/IL-6-stimulated STAT1/STAT3 signaling in mock 22Rv1 cells,
which lack JAK1 expression (Extended Data Fig. 4c-f) suggesting that these pathways are
mediated by endogenous TYK2. Similar to what we observed for JAK1, VVD-118313 inhibited
phosphorylation of the activation loop of WT-, but not C838A-TYK2 (Fig. 3f). BMS-986165 also
suppressed TYK2 phosphorylation; however, this effect was independent of C838 (Fig. 3f).
These data support that VVD-118313 can site-specifically inhibit the signaling of TYK2, at least
We next evaluated the activity of VVD-118313 (5a) and its mixture of stereoisomers (compound
5) in primary human immune cells and found that both compounds inhibited IFNα-pSTAT1, IL-6-
PBMCs (Fig. 2d, e) and substantially blocked IFNα, IL-6, and IL-2 signaling (Fig. 4a-c) - no
concentration than that required to fully engage JAK1_C817, we interpret the modest inhibitory
several cysteines in the human PBMC proteome were substantially engaged by this compound
at 10 µM (Extended Data Fig. 5a and Supplementary Dataset 1). VVD-118313 showed the
11
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
inhibition at 0.01 and 0.1 µM, respectively), followed by IL-6-pSTAT3 (~80-85% inhibition at 0.1-
1 µM) and IL-2-pSTAT5 (~70% inhibition at 0.1-1 µM) pathways. Compound 5 behaved similarly
to VVD-118313, with the expected reduction in potency (Fig. 4a-d) that is in accordance with
TE50 values measured by MS-ABPP (Table 1). The pan-JAK inhibitor tofacitinib blocked all of
found that VVD-118313 did not block TYK2-dependent IL-12-STAT4 signaling in human PBMC-
derived T-blasts, which was inhibited by both BMS-986165 and tofacitinib (Fig. 4e). This result
differed from the inhibitory activity displayed by VVD-118313 in JAK1-null 22Rv1 cells, where
the compound suppressed TYK2-dependent STAT1 phosphorylation and suggests that under
more physiological settings, VVD-118313 does not act as a functional antagonist of TYK2.
We were also interested in evaluating whether the covalent allosteric inhibitors were
capable of engaging and inhibiting JAK1 in vivo. We first confirmed by MS-ABPP that VVD-
118313 selectively and completely engaged C816 of mouse JAK1 (the corresponding residue to
(Extended Data Fig. 5b and Supplementary Dataset 1). Across >11,000 quantified sites, only
a single additional cysteine BACH1_C438 was engaged > 70% by 1 µM of VVD-118313. Mouse
TYK2 was not targeted by VVD-118313 because this protein possesses a serine residue (S858)
(Extended Data Fig. 6a, b). In contrast, VVD-118313 produced only minimal effects (< 30%) on
(Extended Data Fig. 6c, d). One unexpected observation was that VVD-118313 did not inhibit
IL-6-STAT3 signaling in mouse splenocytes (Extended Data Fig. 6e), which contrasted with the
12
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
robust inhibition of this pathway observed in human PBMCs (Fig. 4b). Since tofacitinib retained
inhibitory activity against IL-6-STAT3 signaling in mouse splenocytes (Extended Data Fig. 6e,
h), we speculate that this cytokine pathway may be regulated by different JAK subtypes in
human PBMCs versus mouse splenocytes. In support of this hypothesis, we found that BMS-
greater degree than the pan-JAK inhibitors tofacitinib and upadacitinib (Extended Data Fig. 6f),
suggesting that TYK2 may contribute more substantially than JAK1 to IL-6-stimulated STAT3
We next performed in vivo studies using compound 5, which was chosen over VVD-
118313 due to the comparative ease of scaled-up synthesis and the comparable functional
activity of the mixture of stereoisomers in primary immune cells (Fig. 4a-d). Initial
pharmacokinetic studies revealed that compound 5 exhibited a short half-life (0.36 h) and rapid
clearance in mice (112 mL/min/kg) (Extended Data Table 1). Nonetheless, we hypothesized
that the covalent mechanism of action of 5 may overcome its suboptimal pharmacokinetic
properties to still allow for substantial engagement of JAK1_816 in vivo. The compound was
protocol with a 4 h interval between doses. At 4 h after the second dose, mice were sacrificed
and spleen tissue analyzed by targeted MS-ABPP, which confirmed ~75% engagement of
JAK1_C816 at both 25 and 50 mg/kg of compound 5, while other cysteines in JAK1 were
unaffected (Fig. 4f, g). We also found that splenocytes from compound 5-treated mice showed
from vehicle-treated mice (Fig. 4h and Extended Data Fig 7). In contrast, IL-2-dependent
cultured human (Fig. 4c) and mouse (Extended Data Fig. 6b) immune cells, was not
substantially altered in splenocytes from compound 5-treated mice (Fig. 4h and Extended Data
13
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
Fig 7), suggesting that insufficient JAK1 engagement occurred in vivo to impact this pathway.
Finally, consistent with our cultured immune cell studies, IL-6-STAT3 and GM-CSF-STAT5
signaling were unaffected in splenocytes isolated from compound 5-treated mice (Fig. 4h and
Taken together, our data indicate that covalent ligands engaging human JAK1_C817 (or
mouse immune cells and can serve as chemical probes for both cellular and in vivo studies.
compared to orthosteric inhibitors (Fig. 3c) pointed to distinct mechanisms of action for each
class of compounds. Also consistent with this premise, VVD-118313 did not inhibit the catalytic
activity of recombinant purified JAK1 (aa 438 - 1154, J01-11G, SignalChem) in a peptide
substrate assay (VA7207, Promega), whereas tofacitinib displayed a strong inhibitory effect
(Fig. 5a). We next explored the potential mechanistic basis for VVD-118313 blockade of JAK1
C817A/K908E) variants of JAK1. We first found that, in the absence of VVD-118313, WT-JAK1-
in 22Rv1 cells, while the K908E-JAK1-HA and C817A/K908E-JAK1-HA variants did not show
evidence of phosphorylation (Fig. 5b). Co-expression with either catalytically active JAK1-FLAG
variant (WT or C817A) led to clear trans-phosphorylation of either inactive JAK1-HA variant
in cells co-expressing both C817A-JAK1-FLAG and C817A/K908E-JAK1-HA (Fig. 5b, c). VVD-
14
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
(K908E or C817A/K908E) in cells expressing active WT-JAK1-FLAG, but did not affect trans-
(Fig. 5b, d). We also found that BMS-986165, which has been shown to bind the JAK1 JH2
domain in vitro (IC50 ~ 1 nM) and inhibit JAK1-dependent cytokine signaling (IL-6 and IL-2) with
(Extended Data Fig. 8a). We interpret these data to indicate that the inhibition of trans-
that this effect required the presence of C817 on the donor (phosphorylating), but not the
recipient (phosphorylated) JAK1 variant. That the C817A mutation, on its own, partly
suppressed trans-phosphorylation (Fig. 5b, c), despite not impairing cytokine-induced STAT
phosphorylation (Fig. 3c) can be further interpreted to suggest: i) the potential for allosteric
communication between the C817 pocket of the JH2 domain and the JH1 kinase domain of
JAK1, even in the absence of exogenous inhibitor; and ii) only a modest fraction of
phosphorylated JAK1 is required to fully support cytokine-dependent signaling in the 22Rv1 cell
(e.g., STATs), JAK kinases have also been found to play scaffolding roles in these signal
transduction pathways. For instance, both JAK1 and JAK2 participate in the IFNγ-STAT1
pathway, but only the catalytic activity of JAK2 is required for STAT1 phosphorylation, while the
IFNγ promoted STAT1 phosphorylation in 22Rv1 cells expressing recombinant WT-, K908E-, or
15
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
STAT1 phosphorylation was only supported by catalytically active WT-JAK1, but not the K908E
or C817A/K908E mutants (Fig. 5e). We found that VVD-118313 produced an ~40% partial
C817A/K908E JAK1-expressing cells (Fig. 5f). In contrast, tofacitinib (1 μM, 2h) fully inhibited
C817A/K908E-JAK1 (Fig. 5f), which presumably reflects the blockade of endogenous JAK2
activity by this pan-JAK inhibitor. We further evaluated the impact of VVD-118313 on JAK1
which revealed that upadacitinib blocked all three pathways with similar efficacy (> 90%) and
potency (IC50 values of ~0.10-0.18 µM), while VVD-118313 fully blocked IFNα-mediated
phosphorylation of STAT1 with an IC50 value of ~0.02 µM, partially blocked IFNγ-mediated
phosphorylation of STAT1 with an Imax of ~50%, and did not inhibit GM-CSF-mediated
in human PBMCs. Consistent with our other results (Fig. 4a-d), VVD-118313, at both test
concentrations (0.1 and 1 µM), produced a near complete blockade of JAK1-dependent IFNα-
STAT1 and IL-6-STAT3 signaling and a partial inhibition of IL-2-STAT5 signaling, while showing
negligible effects on JAK2-dependent GM-CSF-STAT5 signaling (Fig. 5h). This profile was
differentiated from the other tested JAK inhibitors – tofacitinib, upadacitinib or itacitinib – all of
which showed weaker potency than VVD-118313 in the IFNα-STAT1 and IL-6-STAT3 signaling
assays, but much greater activity in the IL-2-STAT5 and GM-CSF-STAT5 assays (Fig. 5h and
Extended Data Fig. 8c). The allosteric TYK2 inhibitor BMS-986165 displayed its greatest
16
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
STAT5 signaling, while being inactive against GM-CSF-STAT5 signaling (Fig. 5h and Extended
Data Fig. 8c). Finally, we attempted to measure the effects of VVD-118313 and other JAK
inhibitors on JAK1 phosphorylation, but the signals were too low to visualize in human PBMCs.
Taken together, our studies in primary immune cells illuminate a unique pharmacological
profile of allosteric covalent ligands engaging JAK1_C817 compared to other JAK inhibitors that
includes: i) the strong blockade of cytokine pathways, like IFNα-STAT1 and IL-6-STAT3
signaling, that depend on the catalytic functions of JAK1; ii) the partial inhibition of cytokine
pathways that depend on the scaffolding functions of JAK1 (IFNγ-STAT1) or the activity of
multiple JAK subtypes (IL-2-STAT536); and iii) the sparing of cytokine pathways that depend
(TYK2)).
Discussion
Despite the potential benefits afforded by allosteric over orthosteric kinase inhibitors, which
include not only improvements in selectivity due to interactions with less conserved pockets, but
also avoidance of direct ATP competition for binding, the identification of ligandable and
therefore, may not be detected in more conventional high-throughput assays with purified
kinases and simple peptide substrates, especially if these assays only use truncated catalytic
domains59. Existing allosteric kinase inhibitors have largely been discovered serendipitously or
with detailed knowledge of endogenous regulatory mechanisms56–58. Examples include the “type
III” inhibitors of MEK1/260, LIMK61, and AKT62, which bind to a pocket adjacent to the ATP-
binding site, and the “type IV” inhibitors of Bcr-Abl45,46,63,64, MAPKs65, and receptor tyrosine
17
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
kinases66,67, which bind to allosteric sites distal to the ATP-binding pocket. We have shown here
that chemical proteomics offers a distinct way to discover allosteric inhibitors of kinases.
ligandability of a cysteine that is found in the JH2 pseudokinase domain of JAK1 (C817) and
TYK2 (C838), but not in JAK2 or JAK3. This insight was then efficiently progressed to inhibitory
chemical probes by the coordinated use of targeted chemical proteomic and cell-based
engages and inhibits JAK1 in primary immune cells with low-nanomolar potency, while showing
at a concentration < 1 µM, VVD-118313 had a more subtle functional impact on this kinase, as
we did not observe inhibition of TYK2-dependent cytokine signaling by this compound in human
immune cells. These data indicate that VVD-118313 acts as a functionally selective JAK1
targeting JAK1 while sparing JAK2. As JAK3 only forms heterodimers with JAK1, we did not
assess whether VVD-118313 independently inhibits JAK3 activity in cells, but we do not
anticipate cross-reactivity with JAK3 because it lacks the corresponding liganded cysteine.
Our initial mechanistic studies indicate that VVD-118313 may inhibit JAK1 by blocking
trans-phosphorylation of the activation loop of this kinase. This effect was much stronger for
VVD-118313 compared to orthosteric JAK inhibitors, and we even observed some attenuation
of JAK1 transphosphorylation for the C817A mutant. Our data thus point to a strong potential for
this pocket mirrors the myristate-binding pocket of ABL45–47, it is tempting to speculate that
endogenous metabolites might also bind to JAK1 at this site to regulate its function. We also
wonder how many additional kinases may possess this ligandable pocket and prove amenable
18
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
alignment of 497 human kinase domains reveals a diversity of cysteines at various positions
across more than 14,000 quantified cysteines in human and mouse immune cell proteomes
supports the broader utility of this compound as a cellular probe to investigate the specific
biological functions of JAK1. Indeed, using VVD-118313, we discovered that JAK1 makes
a finding that may have been obscured in past experiments with other JAK1 inhibitors due to
their lack of subtype selectivity. We also found that the TYK2 inhibitor BMS-986165 was
human PBMCs (Extended Data Fig. 6f and Fig. 5h). Thus, by using a combination of allosteric
inhibitors with high subtype selectivity, we have provided evidence for species and/or immune
cell type differences in the relative contributions of JAK family members to an important cytokine
signaling pathway. Curiously, splenocytes from TYK2–/– mice have been reported to have
unperturbed IL-6-STAT3 signaling69,70, which could indicate that, in this setting, JAK1
compensates for chronic TYK2 disruption. VVD-118313 should also help to illuminate JAK1
Projecting forward, we believe that, while VVD-118313 was capable of inhibiting JAK1 in
mice, the full utility of this chemical probe for in vivo studies would benefit from improvements in
its pharmacokinetic properties. We also wonder if further exploration of the SAR might uncover
compounds that show greater functional activity for TYK2, which could provide an additional
class of useful chemical probes that act as dual allosteric JAK1/TYK2 inhibitors. From a
translational perspective, it is enticing to consider the possibility that covalent allosteric JAK1
inhibitors may circumvent some of the systemic toxicities associated with pan-JAK inhibition in
19
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
humans. Alternative ways to address this problem have been brought forward, including tissue-
restricted JAK inhibitors, such as gut-restricted izencitinib (TD-1473) for ulcerative colitis74, and
lung-restricted nezulcitinib (TD-0903) for COVID-1975; however, these compounds have not yet
displayed efficacy in clinical studies in humans76,77, pointing to the potential need for systemic
exposure. Of course, it is possible that selective allosteric JAK1 inhibitors may also sacrifice a
proportion of the efficacy observed with systemic pan-JAK inhibitors, but we believe our cellular
studies showing that VVD-118313 matches the activity of pan-JAK inhibitors in suppressing
multiple human cytokine pathways (e.g., IFNa, IL-6) should encourage further pursuit of highly
Finally, we believe that our findings provide another compelling example of the utility of
chemical proteomics for the discovery of small molecules that act by unconventional
evaluating compounds against thousands of sites on endogenously expressed proteins and can
thus uncover ligandable pockets that may be missed by more conventional assays performed
with purified proteins or protein domains. Nonetheless, chemical proteomics is still principally a
binding assay and interpreting how newly discovered small-molecule interactions affect the
functions of proteins can be technically challenging. Here, we benefited from the availability of
robust cell-based activity assays for JAK1 and, in particular, structural information that
emphasized the potential functionality of a conserved pocket adjacent to the covalently liganded
C817 residue47. As the structures of more full-length proteins are solved or accurately
predicted83,84, the integration of this information with global small-molecule interaction maps
furnished by chemical proteomics should facilitate the discovery of additional cryptic functional
Acknowledgements
20
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
This work was supported by the N.I.H. (R35 CA231991) and a Sir Henry Wellcome Postdoctoral
Wu, X. Jia and M. Xu for their contribution to the synthetic chemistry and B. Melillo for guidance
References
1. Schwartz, D. M. et al. JAK inhibition as a therapeutic strategy for immune and inflammatory
2. Hammarén, H. M., Virtanen, A. T., Raivola, J. & Silvennoinen, O. The regulation of JAKs in
cytokine signaling and its breakdown in disease. Cytokine 118, 48–63 (2019).
3. O’Shea, J. J. et al. The JAK-STAT pathway: impact on human disease and therapeutic
4. Spinelli, F. R., Meylan, F., O’Shea, J. J. & Gadina, M. JAK inhibitors: Ten years after. Eur. J.
5. Clark, J. D., Flanagan, M. E. & Telliez, J.-B. Discovery and development of janus kinase
(JAK) inhibitors for inflammatory diseases. J. Med. Chem. 57, 5023–5038 (2014).
7. Gonzales, A. J. et al. Oclacitinib (APOQUEL®) is a novel Janus kinase inhibitor with activity
against cytokines involved in allergy. J. Vet. Pharmacol. Ther. 37, 317 (2014).
21
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
9. Flanagan, M. E. et al. Discovery of cp-690,550: a potent and selective Janus kinase (JAK)
inhibitor for the treatment of autoimmune diseases and organ transplant rejection. J. Med.
regulation of cytokine signaling in human leukocyte subpopulations. Arthritis Res Ther 21,
183 (2019).
11. Virtanen, A. T., Haikarainen, T., Raivola, J. & Silvennoinen, O. Selective JAKinibs:
candidate for the treatment of autoimmune diseases. J. Med. Chem. 61, 1130–1152 (2018).
13. JAK inhibitors: fate in doubt for rheumatoid arthritis? Editorial. The Lancet Rheumatology 3,
e161 (2021).
14. Baxter, E. J. et al. Acquired mutation of the tyrosine kinase JAK2 in human
15. FDA requires warnings about increased risk of serious heart-related events, cancer, blood
clots, and death for JAK inhibitors that treat certain chronic inflammatory conditions. Feb. 4,
2021. https://www.fda.gov/drugs/drug-safety-and-availability/fda-requires-warnings-about-
1, 2021 (2021).
16. Traves, P. G. et al. JAK selectivity and the implications for clinical inhibition of
17. Choy, E. H. Clinical significance of Janus kinase inhibitor selectivity. Rheumatology 58,
953–962 (2019).
22
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
18. Xu, H. et al. Pf-06651600, a dual jak3/tec family kinase inhibitor. ACS Chem. Biol. 14,
1235–1242 (2019).
19. Tan, L. et al. Development of selective covalent janus kinase 3 inhibitors. J. Med. Chem. 58,
6589–6606 (2015).
21. Wrobleski, S. T. et al. Highly selective inhibition of tyrosine kinase 2 (tyk2) for the treatment
11061–11074 (2015).
23. Abduelmula, A. & Gooderham, M. J. TYK2 inhibition: changing the treatment landscape for
24. Ghoreschi, K. et al. TYK2 inhibition and its potential in the treatment of chronic inflammatory
inhibitor of JAK1, for the treatment of inflammatory diseases. Eur. J. Pharmacol. 885,
173505 (2020).
JAK1 kinase transcriptional and biochemical function. Immunity 53, 672–684 (2020).
28. Mullighan, C. G. et al. JAK mutations in high-risk childhood acute lymphoblastic leukemia.
23
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
29. Li, Q. et al. Identification of a novel functional JAK1 S646P mutation in acute lymphoblastic
30. Ren, Y. et al. JAK1 truncating mutations in gynecologic cancer define new role of cancer-
31. Eletto, D. et al. Biallelic JAK1 mutations in immunodeficient patient with mycobacterial
32. Rodig, S. J. et al. Disruption of the jak1 gene demonstrates obligatory and nonredundant
roles of the jaks in cytokine-induced biologic responses. Cell 93, 373–383 (1998).
33. Kleppe, M. et al. Jak1 integrates cytokine sensing to regulate hematopoietic stem cell
function and stress hematopoiesis. Cell Stem Cell 21, 489–501 (2017).
34. Sakamoto, K., Wehde, B. L., Rädler, P. D., Triplett, A. A. & Wagner, K.-U. Generation of
Janus kinase 1 (JAK1) conditional knockout mice. Genesis 54, 582–588 (2016).
35. Witalisz-Siepracka, A. et al. Loss of JAK1 drives innate immune deficiency. Frontiers in
Immunology 9, (2019).
36. Haan, C. et al. Jak1 has a dominant role over jak3 in signal transduction through γc-
37. Dunn, G. P., Sheehan, K. C. F., Old, L. J. & Schreiber, R. D. IFN unresponsiveness in
LNCaP cells due to the lack of JAK1 gene expression. Cancer Res 65, 3447–3453 (2005).
38. Müller, M. et al. The protein tyrosine kinase JAK1 complements defects in interferon-α/β and
39. Guschin, D. et al. A major role for the protein tyrosine kinase JAK1 in the JAK/STAT signal
transduction pathway in response to interleukin-6. The EMBO Journal 14, 1421 (1995).
40. Telliez, J.-B. et al. Discovery of a jak3-selective inhibitor: functional differentiation of JAK3-
selective inhibition over pan-JAK or JAK1-selective inhibition. ACS Chem. Biol. 11, 3442–
3451 (2016).
24
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
42. Hammarén, H. M. et al. ATP binding to the pseudokinase domain of JAK2 is critical for
43. Shan, Y. et al. Molecular basis for pseudokinase-dependent autoinhibition of JAK2 tyrosine
44. Raivola, J. et al. Hyperactivation of oncogenic JAK3 mutants depend on ATP binding to the
45. Nagar, B. et al. Structural basis for the autoinhibition of c-Abl tyrosine kinase. Cell 112, 859–
871 (2003).
46. Hantschel, O. et al. A myristoyl/phosphotyrosine switch regulates c-abl. Cell 112, 845–857
(2003).
activation of Jak kinases. Nat Struct Mol Biol 20, 1221–1223 (2013).
48. Wylie, A. A. et al. The allosteric inhibitor ABL001 enables dual targeting of BCR–ABL1.
49. FDA approves asciminib for Philadelphia chromosome-positive chronic myeloid leukemia.
approves-asciminib-philadelphia-chromosome-positive-chronic-myeloid-leukemia (accessed
50. Varland, S., Osberg, C. & Arnesen, T. N-terminal modifications of cellular proteins: The
enzymes involved, their substrate specificities and biological effects. Proteomics 15, 2385
(2015).
51. Rostovtsev, V. V., Green, L. G., Fokin, V. V. & Sharpless, K. B. A stepwise Huisgen
25
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
52. Danziger, O., Pupko, T., Bacharach, E. & Ehrlich, M. Interleukin-6 and Interferon-α signaling
via JAK1–STAT differentially regulate oncolytic versus cytoprotective antiviral states. Front.
immunol. 9, 94 (2018).
53. Rossi, M. R. et al. Identification of inactivating mutations in the JAK1, SYNJ2, and CLPTM1
genes in prostate cancer cells using inhibition of nonsense-mediated decay and microarray
gene expression but not an antiviral state. The EMBO Journal 15, 799–809 (1996).
55. Parmentier, J. M. et al. In vitro and in vivo characterization of the JAK1 selectivity of
56. Lu, X., Smaill, J. B. & Ding, K. New promise and opportunities for allosteric kinase inhibitors.
57. Attwood, M. M., Fabbro, D., Sokolov, A. V., Knapp, S. & Schiöth, H. B. Trends in kinase
drug discovery: targets, indications and inhibitor design. Nat Rev Drug Discov 20, 839–861
(2021).
58. Weisner, J. et al. Covalent-allosteric kinase inhibitors. Angew. Chem. Int. Ed. 54, 10313–
10316 (2015).
length HPK1 utilizing a kinase cascade assay. Biochemistry 60, 3114–3124 (2021).
60. Caunt, C. J., Sale, M. J., Smith, P. D. & Cook, S. J. MEK1 and MEK2 inhibitors and cancer
therapy: the long and winding road. Nat Rev Cancer 15, 577–592 (2015).
61. Goodwin, N. C. et al. Discovery of a Type III Inhibitor of LIM Kinase 2 that binds in a DFG-
62. Wu, W.-I. et al. Crystal structure of human AKT1 with an allosteric inhibitor reveals a new
26
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
63. Adrián, F. J. et al. Allosteric inhibitors of BCR-ABL–dependent cell proliferation. Nat Chem
64. Schoepfer, J. et al. Discovery of asciminib (ABL001), an allosteric inhibitor of the tyrosine
65. Comess, K. M. et al. Discovery and characterization of non-ATP site inhibitors of the
Mitogen Activated Protein (MAP) kinases. ACS Chem. Biol. 6, 234–244 (2011).
67. Bono, F. et al. Inhibition of tumor angiogenesis and growth by a small-molecule multi-FGF
receptor blocker with allosteric properties. Cancer Cell 23, 477–488 (2013).
70. Minegishi, Y. et al. Human Tyrosine Kinase 2 deficiency reveals its requisite roles in multiple
cytokine signals involved in innate and acquired immunity. Immunity 25, 745–755 (2006).
71. Neilson, L. M. et al. Coactivation of Janus tyrosine kinase (Jak)1 positively modulates
prolactin-Jak2 signaling in breast cancer: recruitment of Eek and signal transducer and
activator of transcription (Stat)3 and enhancement of Akt and Stat5a/b pathways. Mol.
72. Huang, F. et al. Requirement for both JAK-mediated PI3K signaling and
27
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
73. Friedmann, M. C., Migone, T. S., Russell, S. M. & Leonard, W. J. Different interleukin 2
receptor beta-chain tyrosines couple to at least two signaling pathways and synergistically
74. Sandborn, W. J. et al. Development of gut-selective pan-Janus kinase inhibitor TD-1473 for
(2020).
75. Pfeifer, N. D., Lo, A., Bourdet, D. L., Colley, K. & Singh, D. Phase I study in healthy
potential treatment for COVID-19. Clin. Transl. Sci. 14, 2556–2565 (2021).
76. Theravance’s nezulcitinib fails to meet Phase II goals in Covid-19. June 21, 2021.
https://www.clinicaltrialsarena.com/news/theravance-nezulcitinib-phaseii-data/ (acessed
77. Theravance’s izencitinib fails in Phase IIb ulcerative colitis trial, Aug 24, 2021.
https://www.clinicaltrialsarena.com/news/theravance-izencitinib-ulcerative-colitis/ (accessed
78. Backus, K. M. et al. Proteome-wide covalent ligand discovery in native biological systems.
80. Boike, L. et al. Discovery of a functional covalent ligand targeting an intrinsically disordered
81. Spradlin, J. N. et al. Harnessing the anti-cancer natural product nimbolide for targeted
82. Chung, C. Y.-S. et al. Covalent targeting of the vacuolar H+-ATPase activates autophagy
28
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
83. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596,
583–589 (2021).
84. Tunyasuvunakool, K. et al. Highly accurate protein structure prediction for the human
29
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
30
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
31
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
32
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
33
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
ng/mL, 30 min), IL-6 (50 ng/mL, 30 min) or PRL (15 ng/mL, 15 min) prior to analysis. c, d,
Quantification of pSTAT1/3/5 (c) and pJAK1 (d) signals from (b). Signal intensities were
normalized relative to the unstimulated WT-JAK1-transfected control for each experiment. Data
are mean values ± S.E.M. from three independent experiments. Significance was determined by
two-way ANOVA with Dunnett’s post-hoc test and reported relative to stimulated, DMSO-treated
control of respective JAK1 construct. ****P<0.0001, ***P<0.001, **P<0.01, *P<0.05. e,
Concentration-dependent effects of VVD-118313 (5a) on IFNα-stimulated pSTAT1 (left), IL-6-
stimulated pSTAT3 (middle), and pJAK1 (integrated from both IFNα- and IL-6-stimulations) in
22Rv1 cells expressing WT-JAK1. Data are mean values ± SEM from two (pSTAT1, pSTAT3)
or three (pJAK1) independent experiments. See Extended Data Fig. 3b, c for corresponding
western blots. f, Concentration-dependent effects of VVD-118313 (5a; 0.01 – 5 µM, 2 h) and
BMS- 986165 (BMS, 1 or 5 µM, 2 h) on TYK2 phosphorylation (pTYK2) and IFNα-stimulated
STAT1 phosphorylation in 22Rv1 cells expressing recombinant WT-TYK2 or a C838A-TYK2
mutant. Left, representative western blots. Middle and right, quantification of pSTAT1 (middle)
and pTYK2 (right) signals normalized to unstimulated control cells expressing WT-TYK or
C838A-TYK2. Data are mean values ± S.E.M from three independent experiments. Significance
was determined by two-way ANOVA with Dunnett’s post-hoc test. P-values are only shown for
the lowest concentration of each compound that displayed significance for inhibition of pSTAT1
and pTYK2. ****P<0.0001, ***P<0.001.
34
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
Figure 4. VVD-118313 selectively inhibits JAK1 signaling in primary human immune cells
and mice. a-d, Effects of VVD-118313 (5a), stereoisomeric mixture 5, and tofacitinib (Tofa) on
JAK-STAT signaling pathways in human PBMCs. PBMCs were treated with compounds at the
indicated concentrations for 2 h prior to stimulation with IFNα (a; 100 ng/mL, 30 min), IL-6 (b; 25
ng/mL, 30 min), IL-2 (c; 20 U/mL, 15 min), or GM-CSF (d; 0.5 mg/mL, 15 min). Upper,
representative western blots. Lower, Quantification of pSTAT signal intensities shown as a
percent of the stimulated DMSO-treated control cells for each assay. Data are mean values ±
S.E.M. from three (IL-6, IL-2) or four (IFNα, GM-CSF) independent experiments. Significance
determined by one-way-ANOVA with Dunnett’s post-hoc test. P-values are only shown for the
lowest concentration of each compound that displayed significance for inhibition of pSTAT. All
higher concentrations were similarly statistically significant. ****P<0.0001, ***P<0.001, **P<0.01,
*P<0.05. e, Effects of VVD-118313 (5a), compound 5, BMS-986195 (BMS), and tofacitinib
(Tofa) on IL-12-stimulated STAT4 phosphorylation in phytohemagglutinin (PHA-P)/IL-2-
generated PBMC-derived T-blasts. Cells were treated with compounds at the indicated
concentrations for 2 h prior to stimulation with IL-12 (12.5 ng/mL, 15 min). Data are mean values
35
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
± S.E.M. normalized to DMSO controls from three independent experiments and significance
was determined as for (a-d). f, g, Reactivity profiles for JAK1_C816 (f) and all quantified JAK1
cysteines (g) from proteomic lysates of spleen tissue from mice treated with vehicle (dose 0
mg/kg) or compound 5 (25 or 50 mg/kg, s.c. 2 x 4 h). Data are mean values ± S.D. from
compound 5-treated mice shown as a percentage of vehicle-treated mice (n = 4 animals/group
analyzed in a single targeted MS-ABPP). In g, bars represent median reactivity values for all
JAK1 cysteines other than C816. Significance determined by one-way ANOVA with Dunnett’s
post hoc test. ****P<0.0001. h, Ex vivo cytokine stimulation of splenocytes from mice treated
with vehicle or 5 (25 mg/kg, s.c., 2 x 4 h); IFNα (100 ng/mL, 30 min), IL-2 (20 U/mL, 15 min), IL-
6 (10 ng/mL, 30 min) or GM-CSF (10 ng/mL, 15 min). Data are mean values ± S.E.M., from
three (IFNα, IL-2) or one (IL-6, GM-CSF) independent experiments, each containing n = 3 mice
per treatment group. Significance determined by two-way ANOVA with Šidák’s post hoc test.
****P<0.0001.
36
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
Figure 5. Mechanistic properties and distinct activity profile of allosteric JAK1 inhibitors.
a, Substrate assay quantifying ATP turnover by recombinant purified JAK1 (residues 438-1154
fused to GST) treated with DMSO, VVD-118313 (5a) or tofacitinib (Tofa) (0.001 - μM, 30 min)
prior to addition of an IRS-1 peptide substrate (0.2 µg/mL) and ATP (50 µM, 1 h). Data are
mean values ± S.D. from two independent experiments. b, Western blots measuring JAK1
phosphorylation (pJAK1) from anti-HA immunoprecipitations (IPs) of HA-tagged kinase dead
(K908E) JAK1 (WT or C817A mutant) expressed in 22Rv1 cells alongside catalytically active
FLAG-tagged JAK1 (WT or C817A mutant). c, d, Quantification of pJAK1 signals from anti-HA-
IPs shown in b. Panel c shows pJAK1 signals from anti-HA IPs from DMSO-treated cells co-
expressing the indicated combinations of K908E-JAK1-HA or C817A/K908E-JAK1-HA with WT-
37
bioRxiv preprint doi: https://doi.org/10.1101/2022.01.31.478302; this version posted February 1, 2022. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY-NC-ND 4.0 International license.
JAK1-FLAG or C817A-JAK1-FLAG. Panel d shows pJAK1 signals for the same anti-HA IPs
from VVD-118313 (5a; 2 µM, 2 h)-treated 22Rv1 cells. Data in c are normalized to signals in
22Rv1 cells expressing K908E-JAK1-HA and WT-JAK1-FLAG. Data in d are normalized to
signals in DMSO-treated control cells Data for c and d are mean values ± S.E.M. from three
independent experiments. Significance was determined by two-way ANOVA with Dunnett’s
post-hoc test. ***P<0.001, ****P<0.0001. e, Upper, western blots showing that both K908E- and
K908E/C817A-JAK1 mutants support IFNγ-stimulated (50 ng/mL, 30 min), but not IFNα-
stimulated (100 ng/mL, 30 min) STAT1 phosphorylation (pSTAT1) in 22Rv1 cells. WT-JAK1
supports both cytokine pathways. Lower, quantification of western data. Data are mean values ±
S.E.M. from three independent experiments. pSTAT1 signals were normalized to the maximum
signal, which was generated by IFNγ-stimulated WT-JAK1 transfected cells. Significance was
determined by two-way ANOVA with Tukey’s post-hoc test. ****P<0.0001, ***P<0.001. ns – no
significance difference between IFNα-stimulated K908E-JAK1-HA- or K908E/C817A-JAK1-HA-
expressing cells and mock cells. f, Left, western blots showing the effects of VVD-118313 (5a;
0.1-5 µM, 2 h) and tofacitinib (Tofa; 1 µM, 2 h) on IFNγ-dependent STAT1 phosphorylation
(pSTAT1) in 22Rv1 cells expressing K908E-JAK1-HA or K908E/C817A-JAK1-HA. Right,
quantification of western data. Data are mean values ± S.E.M. from three independent
experiments. Significance determined by two-way ANOVA with Dunnett’s post-hoc test.
*P<0.05, **P<0.01, ****P<0.0001, ns – non-significant. g, Concentration-dependent effects of
VVD-118313 (5a) or upadacitinib (Upa) on IFNα-dependent STAT1, IFNγ-dependent STAT1,
and GM-CSF-dependent STAT5 phosphorylation in human PBMCs. pSTAT signals were
normalized to the DMSO-treated cytokine-stimulated control in each assay. Dose-response
curves are mean values ± S.D. of two biological replicates used to estimate IC50 values by
fitting data to a 4PL model. h, Left, western blots comparing the effects of VVD-118313 (5a) and
a panel of JAK inhibitors on the indicated cytokine-stimulated pSTAT pathways in human
PBMCs. Right, quantification of western data. Data are mean values ± S.E.M. from two (IL-6) or
three (IFNα, IL-2, GM-CSF) independent experiments. Significance determined by one-way
ANOVA with Šidák’s post-hoc test. *P<0.05, ****P<0.0001, ns – non-significant
38