The Society for Immunotherapy of Cancer (SITC) is pleased to present scientific highlights from the Nov. 10, 2018, sessions of the 33rd Annual Meeting.
![]() |
Kristin Anderson, PhD (Fred Hutchinson Cancer Research Center), delivers her SITC 2018 Presidential Session presentation on Saturday, Nov. 10, 2018. |
The Novel Oncolytic Virus Voyager V1 is Safe and Promotes Immunity in Patients with Solid Tumors
Steven F. Powell, MD (Sanford Health, Sioux Falls, SD, USA)
detailed first-in-human data for application of the novel oncolytic virus
Voyager V1 (VV1) for treatment of patients with solid tumors including squamous
cell carcinoma of the head and neck, colon, rectal, pancreatic, and breast,
among other cancers. VV1 is derived from the vesicular stomatitis virus and
encodes IFNbeta to help amplify anti-tumor efficacy, as well as the thyroidal
sodium iodide symporter NIS gene to allow for noninvasive imaging of virus
dissemination. Administered as a one-time intratumoral injection, VV1 was well
tolerated in patients with 80% of treatment related AEs reported being
short-lived as well as mild to moderate in severity. Interestingly, VV1
proliferation and spread, determined by positive SPECT/CT within the tumor, was
more readily observed in colorectal and pancreatic cancers compared to other
diseases. Additionally, IFNbeta was detected in the serum of patients treated
with high VV1 doses, indicative of viral replication within the tumor.
Importantly, there was evidence of intratumoral inflammation in 75% of
evaluable patients, and T cell infiltration was observed up to 29 days
post-therapy in non-injected lesions. Overall, these data indicate that VV1 is
well tolerated as a single-agent, and that it may promote inflammation and
immunity at both injected and non-injected tumors. Having demonstrated
synergistic effects with anti-PD-1/PD-L1 antibodies in preclinical models,
future evaluations of VV1 in combination with the anti-PD-L1 avelumab are
planned.
Chop Directly Regulates T-bet Transcription to Control T cell Motility
Yu Cao, PhD (H. Lee Moffitt Cancer Center, Tampa, FL, USA)
discussed research evaluating the role of C/EBP-homologous protein Chop in the regulation
of CD8+ T cell effector function in the tumor microenvironment. Data indicate
that Chop is upregulated in tumor-associated CD8+ T cells from human ovarian
cancer samples, but not in circulating CD8+ T cells. Furthermore, ovarian
cancer patients with CD8+ T cells expressing high-levels of Chop have reduced
survival compared to patients with Low-Chop expressing intra-tumor CD8+ T cells
(p = 0.05). Next-generation sequencing analyses revealed overexpression of signatures
associated with effector function in Chop-negative CD8+ T cells. Accordingly,
Chop-negative CD8+ T cells displayed increased anti-tumor activity. Further
experiments showed that Chop deletion in T cells results in increased
expression of T-bet – the master regulator of effector T cell function.
Interestingly, a Chop binding site was identified within the promoter region of
T-bet, and subsequent experiments revealed that Chop in fact binds to T-bet
promoter and directly inhibits T-bet transcription. Finally, adoptively
transferred Chop-negative CD8+ T cells more efficiently controlled tumor growth
in B16-bearing mice compared to Chop-positive cells. Together, these data
suggest that Chop directly inhibits T-bet transcription and regulates CD8+ T
cell effector function, offering a new potential target to augment
immunotherapeutic efficacy.
Phase 1b Data Reveal Early Signs of Safety and Efficacy of the A2AR Antagonist CPI-444 as Monotherapy and in Combination with Atezolizumab in Patients with Renal Cell Carcinoma
Lawrence Fong, MD (University of California, San Francisco,
CA, USA) reported data from a phase 1b clinical trial (NCT02655822) evaluating
CPI-444 - an oral antagonist of the adenosine 2A receptor (A2AR) - as
monotherapy or in combination with anti-PD-L1 atezolizumab for the treatment of
patients with relapsed/refractory renal cell carcinoma (RCC). Patients (median three
prior treatments, 71 percent prior-immunotherapy) received either CPI-444
(100mg BID) as a monotherapy (n=33) or in combination with atezolizumab (800 mg
Q2W, n=35). At median follow up of 8.7 months, overall survival was 65% and 88%
for the monotherapy and combination cohorts, respectively. In addition, median
progression-free survival in the monotherapy cohort was 4.1 mos, and 5.8 mos
patients receiving combination CPI-444 + atezolizumab. 6-month disease control
rates (DCR) were 25% and 32% for the monotherapy and combination cohorts,
respectively. Median time to best tumor response was 3.4 months for the
monotherapy cohort and 5.5 months for the combination cohort. CPI-444 was well
tolerated with mostly low-grade treatment-related adverse events. Increased
CD8+ T cell infiltration positively correlated with improved 6-month DCR.
Analyses revealed that clinical response was only seen in patients with high
tumor-adenosine signatures. In summary, data suggest early clinical benefit of
CPI-444 monotherapy or in combination with atezolizumab in patients with
relapsed/refractory RCC, and that tumor adenosine gene signatures may be a
potential biomarker of response.
Targeting FasL using an Immunomodulatory Protein May Help Increase Persistance of Adoptive TCR T cell Therapies
Kristin Anderson, PhD (Fred Hutchinson Cancer Research
Center, Seattle, WA, USA) presented data demonstrating how an immunomodulatory
fusion protein targeting Fas ligand (FasL) can enhance survival of adoptive TCR-transduced
T cells in a pre-clinical ovarian cancer model. In collaboration with Dr.
Philip Greenberg's group, Dr. Anderson utilized adoptive T cells that harbor a
genetically-engineered TCR receptor targeting mesothelin (TCR-Mslin) as a
potential therapeutic to treat patients with ovarian cancer. Results indicate
that TCR-Mslin T cells have anti-tumor activity in vitro, but have limited
persistence and reduced effector function in pre-clinical ID8-VEGF tumors that
mimic human ovarian cancer phenotypes. Multiple adoptive transferes of
TCR-Mslin T cells increased survival of mice bearing ID8-VEGF tumors, but this
strategy has limited translational potential in humans. As such, Anderson’s
group developed a FasL/4-1BB immunomodulatory fusion protein to help increase
CD8+ T cell proliferation and survival. FasL/4-1BB CD8+ T Cells had increased
proliferation ability, as well as increased cytokine secretion including
IFNgamma and TNFalpha. Enhanced survival was noted in mice bearing ID8-VEGF
tumors who received adoptive transfer of TCR-Mslin + FasL/4-1BB T cells when
compared to mice receiving TCR-Mslin or WT CD8+ T cells. In all, these early
data suggest that the described FasL/4-1BB immunomodulatory protein may help
increase survival of TCR-transduced CD8+ T cells, and may be a potential
technology to help facilitate movement of TCR-T cells into the clinic.
PAK4 Regulates Immune Cell Infiltration to Increase Tumor Response to Anti-PD-1 Blockade
Gabriel Abril-Rodriguez (UCLA, Los Angeles, CA, USA)
discussed analyses of tumor samples from patients with metastatic melanoma
treated with anti-PD-1 therapy to help identify mechanisms leading to tumoral
immune cell exclusion. RNAseq analyses performed on tumor samples from 41
melanoma patients demonstrated that low CD8+ T cell and dendritic cell
infiltration an inverse correlation correlate with upregulation of P21
activated Kinase 4 (PAK4). PAK4 is a kinase that binds to and phosphorylates
beta-catenin to activate Wnt signaling, and is known to be involved in
tumorigenesis. This observation was further supported by analyses of TCGA gene expression
data from 32 cancer types showing a negative correlation between PAK4
expression and clinical responses in patients with melanoma, prostate cancer,
and pancreatic cancer, among others. Syngeneic C57BL/6 mice bearing B16 PAK4
knockout tumors were sensitized to PD-1 inhibition compared to mice bearing B16
wildtype tumors that were resistant to checkpoint blockade, and PAK4 knockout
tumors displayed increased CD8+ and dendritic cell infiltration compared to WT
tumors. Anti-PD-1 efficacy was lost upon depletion of CD8+ T cells in PAK4
knockout tumors. Importantly, treatment of B16 melanoma models with anti-PAK4 and
anti-PD-1 significantly reduced tumor growth compared to mice treated with each
agent alone. Together, these data suggest that PAK4 may contribute to tumor
immune evasion, and that inhibition of PAK4 may help overcome resistance to
PD-1 blockade.
B cell Signatures may Predict Response to Neoadjuvant Immune Checkpoint Blockade in Melanoma and Renal Cell Carcinoma Patients
Sangeetha Reddy, MD, MSci (MD Anderson Cancer Center,
Houston, TX, USA) presented research towards the identification of immune cell
profiles of response in patients from a randomized trial (NCT02519322)
evaluating efficacy of neoadjuvant immune checkpoint blockade in patients with
high-risk resectable melanoma. A total of 23 patients were randomized to
receive neoadjuvant nivolumab (n = 12) or neoadjuvant ipilimumab + nivolumab (n
= 11), and tumor samples were analyzed using transcriptomic profiling. Typical
biomarkers associated with response to immune checkpoint inhibitors – including
PD-L1 and CD8 – were observed in patient tumor samples. Interestingly, tumor B
cell signatures were also differentially expressed according to patient
response, including MZB1, JCHAIN, and IGLL5 (p less than 0.0001 for each). B
cell lineage score was also correlated with improved survival, especially in
patients who had low CD8+ T cell signatures in their respective tumor samples.
Interestingly, data from a similar trials evaluating neoadjuvant immune
checkpoint inhibition in patients with metastatic RCC (NCT02210117), as well as
melanoma samples within the TCGA dataset displayed immune signature patterns to
the observed melanoma trial dataset. Multiplex immunohistochemostry of tumor
sections from the melanoma trial cohort demonstrated co-localization of the B
cells and CD8+, CD4+ T cells, as well as CD21 follicular dendritic cells, in
tertiary lymphoid structures or responders (p = 0.037 at baseline, p = 0.002
on-treatment). Together, these results suggest that B cell genetic signatures
may serve as prognostic and predictive biomarkers for response to neoadjuvant
immune checkpoint blockade.
Increased Abundance of CD8+ Stem Memory Cells Correlates with Response in Melanoma Patients Treated with Adoptive TIL Therapy
Matthew Beatty (Moffitt Cancer Center, Tampa, FL, USA)
detailed novel data describing how enrichment of CD8+ stem memory T cells
(Tscm) in adoptive T cell therapy products associates with response in patients
with advanced melanoma. In all, 57 patients with stage IV melanoma (anti-PD-1
naïve) were enrolled across four trials and 47 patients received adoptive therapy
with tumor-infiltrating lymphocytes (TIL). Overall response rate (ORR) in the
treated patients was nearly 39 percent. Interestingly, CD8+ Tscm cell and
overall TIL persistence were increased in patients with complete or partial
response compared to patients who demonstrated stable or progressive disease
(per RECIST). Increased five-year PFS (36.5 months) was observed in patients
with high levels of Tscm (greater than 7.81 percent), compared to patients with
low levels of Tscm (less than 7.81 percent; seven months). Furthermore, T cell
clones derived from infusion product CD8+ Tscm were shown to persist/expand in
vivo at six weeks post-infusion, and there was a positive association between
CD8+ Tscm abundance and one-year clinical response. In addition, purified
patient CD8+ Tscm demonstrated anti-tumor reactivity as well as ability to
differentiate into additional memory T cell subsets six weeks post-infusion. In
summary, CD8+ Tscm abundance in adoptive TIL infusion products may indicate
increased anti-tumor efficacy in patients with advanced melanoma who have not
received prior anti-PD-1 treatment.
IL10 and IL35 may Regulate CD8+ Inhibitory Receptor Expression Through BLIMP1
Hiroshi Yano, BS (University of Pittsburgh, Pittsburgh, PA,
USA) presented research investigating the cooperative regulation of anti-tumor
immunity in the tumor microenvironment by IL35 and IL10. Researchers discovered
that deletion of IL10, IL35, and IL10/IL35 similarly lead to reductions in in
vivo B16-melanoma growth. Interestingly, IL10 and IL35 deficiency individually
reduced CD8+ inhibitory receptor expression – including TIM3, TIGIT, and PD-1.
IFNgamma and TNFalpha secretion, however, were differentially regulated by IL10
and IL35, as well as CD8 memory T cell differentiation. Towards elucidating a
potential mechanism, Yano’s group analyzed CD8+ T cell subsets from IL10 and
IL35 deficient mice and noted increased expression of BLIMP1 – a known
regulator of T cell inhibitory receptor expression. Further experiments
demonstrated that IL10 and IL35 directly inhibit BLIMP1 expression, thus
explaining CD8+ T cell inhibitory receptor down-regulation in IL10 and IL35
deficient mice. Together, these data help describe a mechanism of how IL10 and
IL35 can contribute to CD8+ T cell anti-tumor activity through direct
regulation of BLIMP1 and control of inhibitory receptor expression, but also
differentially regulate cytokine secretion and memory cell expansion.
No comments:
Post a Comment