CRC 24 0082 PDF
CRC 24 0082 PDF
CRC 24 0082 PDF
ABSTRACT
Purpose: TPST-1120 is a first-in-class oral inhibitor of peroxisome ported. In the monotherapy group, 53% (10/19) of evaluable patients had
proliferator-activated receptor α (PPARα), a fatty acid ligand-activated a best objective response of stable disease. In the combination group, 3 pa-
transcription factor that regulates genes involved in fatty acid oxidation, tients had partial responses, for an objective response rate of 20% (3/15)
angiogenesis, and inflammation, and is a novel target for cancer therapy. across all doses and 30% (3/10) at TPST-1120 ≥400 mg twice daily. Re-
TPST-1120 displayed antitumor activity in xenograft models and syner- sponses occurred in 2 patients with RCC, both of whom had previously
gistic tumor reduction in syngeneic tumor models when combined with progressed on anti-PD-1 therapy, and 1 patient with late-line CCA.
anti-PD-1 agents. Conclusions: TPST-1120 was well tolerated as monotherapy and in combi-
Experimental Design: This phase I, open-label, dose-escalation study nation with nivolumab and the combination showed preliminary evidence
(NCT03829436) evaluated TPST-1120 as monotherapy in patients with ad- of clinical activity in PD-1 inhibitor refractory and immune compromised
vanced solid tumors and in combination with nivolumab in patients with cancers.
renal cell carcinoma (RCC), cholangiocarcinoma (CCA), or hepatocellu- Significance: TPST-1120 is a first-in-class oral inhibitor of PPARα, whose
lar carcinoma. Objectives included evaluation of safety, pharmacokinetics, roles in metabolic and immune regulation are implicated in tumor prolifer-
pharmacodynamics, and preliminary antitumor activity (RECIST v1.1). ation/survival and inhibition of anticancer immunity. This first-in-human
Results: A total of 39 patients enrolled with 38 treated (20 monother- study of TPST-1120 alone and in combination with nivolumab sup-
apy, 18 combination; median 3 prior lines of therapy). The most common ports proof-of-concept of PPARα inhibition as a target of therapeutic
treatment-related adverse events (TRAE) were grade 1–2 nausea, fatigue, intervention in solid tumors.
and diarrhea. No grade 4–5 TRAEs or dose-limiting toxicities were re-
Introduction cancer is an increase in aerobic glycolysis (the Warburg effect), cancer cells
can utilize other metabolic pathways such as increased fatty acid oxidation
Metabolic reprogramming and evasion of immune destruction are hallmarks (FAO) to support tumorigenesis. FAO also promotes stemness, drug resistance,
of cancer (1). While the most widely recognized metabolic adaptation in and metastasis (2–4), and modulates immune cell function within the tumor
microenvironment, which enables tumors to evade antitumor immune re- Patient Selection
sponses (5–7). Enhanced FAO is described in multiple cancers and has been
Key inclusion criteria included diagnosis of advanced/metastatic solid tumor
reported to correlate with poor patient outcomes (8).
previously treated with standard systemic therapy for the disease; Eastern
Peroxisome proliferator-activated receptor α (PPARα) is a fatty acid ligand- Cooperative Oncology Group (ECOG) performance status of 0–1 with es-
activated transcription factor that controls the expression of over 100 genes timated life expectancy of at least 12 weeks; adequate organ function; and
involved in FAO, angiogenesis, and inflammation (9–11). PPARα is crit- measurable disease according to the RECIST version 1.1. Tumor types eligi-
ical for maintaining physiologic metabolic homeostasis under conditions ble for monotherapy dose escalation [cholangiocarcinoma (CCA), colorectal
when fatty acids are the predominant source of energy, such as during cancer, metastatic castration-resistant prostate cancer, gastroesophageal can-
fasting or diet-induced lipid overload. In addition to upregulating genes cer, hepatocellular carcinoma (HCC), non–small cell lung cancer, pancreatic
involved in FAO, activated PPARα dampens Th1-promoting inflammatory cancer, renal cell carcinoma (RCC), sarcoma, head and neck squamous cell
responses to metabolic disturbances by directly enhancing transcription of carcinoma, triple-negative breast cancer, and urothelial bladder cancer] were
anti-inflammatory proteins, such as IκBα, and by antagonizing the activity selected based upon elevated expression of PPARA and associated genes
of proinflammatory transcription factors, such as NFκB and AP-1, through in The Cancer Genome Atlas in these indications. In the nivolumab com-
Pharmacokinetics and Pharmacodynamics Overall response rate (ORR) was defined as the proportion of patients with
complete response or partial response (PR). Disease control rate (DCR) was de-
Blood for pharmacokinetic and research assessments was collected at screening,
fined as ORR + SD of at least one scan. The efficacy population (EP) comprised
cycle 1 days 1, 2, and 8, cycle 2 day 1, cycle 3 day 1, cycle 4 day 1 (combination
all safety-evaluable patients who had at least one postbaseline tumor assess-
only), and cycle 5 day 1 (monotherapy only). Tumor biopsies were not required.
ment, as well as patients who discontinued from study treatment due to PD
Plasma TPST-1120 concentrations were quantified using a validated tandem without undergoing a follow-up radiographic assessment.
mass spectrometry assay. Noncompartmental analysis was performed on ob-
served data following the first dose and at steady state on day 8. To assess Study Approval
pharmacodynamic gene expression changes, RNA was extracted from whole
This study was reviewed and approved by the individual site Institutional Re-
blood samples collected in PAXgene Blood RNA tubes on cycle 1 days 1 and 8
view Boards and/or ethics committees where the study was opened and by
and cycle 3 day 1 and assayed on the nCounter instrument (NanoString, Inc.)
the FDA. The study was conducted in accordance with the provisions of the
using the nCounter PanCancer Immune Profiling panel supplemented with an
Declaration of Helsinki and the International Conference on Harmonization
additional 30 PPARα-associated genes (Supplementary Table S1) according to
Guidelines for Good Clinical Practice. All patients provided written informed
DLTs were evaluated during the first treatment cycle (21 days for monother-
Pharmacokinetics
apy and 28 days for TPST-1120 plus nivolumab). Patients were evaluable for
DLTs if they received at least 85% of planned TPST-1120 doses (and all planned Data from 31 patients were available for pharmacokinetic analysis on day 8.
nivolumab doses for combination patients) in the first treatment cycle un- TPST-1120 steady-state exposure levels increased in a linear, dose-dependent
less this exposure threshold was not met due to a DLT. Patients who did manner and were not affected by nivolumab (Supplementary Fig. S1). Key phar-
not meet the DLT evaluability criteria were replaced. AEs occurring dur- macokinetic parameters of patients receiving TPST-1120 at 600 mg following
ing the first treatment cycle and assessed as related to study treatment were single dose and at steady state are listed in Supplementary Table S2.
considered DLTs following criteria established in the study protocol. OBD
determination was based on emerging pharmacokinetics, any pharmacoki- Safety
netic/pharmacodynamic relationships which could be established, and overall No DLTs occurred during dose escalation. Two monotherapy patients (1 at
safety and tolerability. 200 mg twice daily and 1 at 400 mg twice daily) were not evaluable for DLTs
due to missing more than 15% of the required TPST-1120 doses during the eval-
Statistical Methods uation period due to AEs unrelated to treatment and were replaced. An MTD
Analysis was performed using descriptive statistics. All patients who received was not established for TPST-1120 as monotherapy or in combination with
at least one dose of TPST-1120 were included in the safety analysis. nivolumab.
1102 Cancer Res Commun; 4(4) April 2024 https://doi.org/10.1158/2767-9764.CRC-24-0082 | CANCER RESEARCH COMMUNICATIONS
PPARα Inhibition in Patients with Advanced Solid Cancers
TABLE 1 Demographics and patient characteristics of the monotherapy and combination cohorts
TPST-1120 TPST-1120 +
Monotherapy Nivolumab
Baseline characteristics (n = 20) (n = 18)
Abbreviations: BID, twice daily; ECOG PS, Eastern Cooperative Oncology Group performance status.
In the monotherapy cohort, 10 patients (50%) experienced an AE that was was assessed as treatment-related. There were no grade 4 or grade 5 TRAEs,
deemed as related to TPST-1120 (Table 2). The most frequently reported and no patient discontinued study treatment due to a TRAE.
treatment-related AEs (TRAE) were nausea [4 (20%) patients], fatigue [3 (15%)
In the combination therapy cohort, 14 patients (77.8%) experienced TRAEs re-
patients], and diarrhea [2 (10%) patients], all grade 1–2. One patient experi-
lated to either TPST-1120 or nivolumab (Table 2). The most frequent TRAEs
enced grade 3 hypertension at the 600 mg twice daily dose of TPST-1120 that
were fatigue [6 (33.3%) patients], diarrhea [4 (22%) patients], and nausea [3
(17%) patients], all grade 1–2. Three patients experienced grade 3 TRAEs:
1 each of arthralgia (TPST-1120 400 mg twice daily), hepatic enzymes in-
TABLE 2 Treatment-related treatment-emergent AEs by preferred
creased (TPST-1120 600 mg twice daily), and muscle spasms (TPST-1120 600 mg
term in ≥1 patient, any grade and grade 3
twice daily). Two of these TRAEs (grade 3 arthralgia and grade 3 hepatic en-
AE, n (%) Grades 1–3a Grade 3 zymes increased) were considered immune-related and treated with systemic
steroids. There were no grade 4 or grade 5 TRAEs. The patient with grade 3
TPST-1120 Monotherapy (n = 20) hepatic enzymes increased was the sole patient treated with combination ther-
Patients with ≥1 TRAE 10 (50.0) 1 (5.0) apy (TPST-1120 600 mg orally twice daily) to discontinue treatment due to
Nausea 4 (20.0) — a TRAE.
Fatigue 3 (15.0) —
Diarrhea 2 (10.0) —
Efficacy
Hypertension 1 (5.0) 1 (5.0)
Clinical efficacy for TPST-1120 monotherapy and in combination with
TPST-1120 + Nivolumab (n = 18)
nivolumab is summarized in Fig. 1. Four treated patients were not included
Patients with ≥1 TRAEb 14 (77.8) 3 (16.7)
in the EP due to discontinuation of treatment for either symptomatic deteri-
Fatigue 6 (33.3) —
oration without objective evidence of PD (1 monotherapy, 1 combination) or
Diarrhea 4 (22.2) —
due to unrelated AE (2 combination patients) prior to obtaining a postbaseline
Nausea 3 (16.7) —
tumor assessment. The monotherapy efficacy evaluable population included
Abdominal pain 2 (11.1) —
3 patients who did not have a postbaseline RECIST assessment but discon-
Arthralgia 1 (5.6) 1 (5.6)
tinued treatment for investigator-assessed disease progression not confirmed
Hepatic enzyme increased 1 (5.6) 1 (5.6)
by RECIST. Among the 19 efficacy evaluable patients in the monotherapy co-
Muscle spasms 1 (5.6) 1 (5.6)
hort, the BOR was SD for 10 (53.0%) patients and PD for 6 (32.0%) patients,
a No grade 4 or 5 TRAEs. for a DCR of 53%. Tumor shrinkage of target lesions was observed in 4 patients
b Related to either TPST-1120 or nivolumab. (21%) with no target lesion growth as the best relative change from baseline in
combination therapy, scans occurred every 8 weeks. The RCC responder in the 600 mg BID + nivolumab dose group had an unscheduled scan at day
24 during hospitalization for treatment-unrelated AEs. This patient discontinued treatment one day prior to the scan but was unable to restart and had
disease progression during the posttreatment follow-up period. BID, twice daily; CCA, cholangiocarcinoma; CRC, colorectal carcinoma; HCC,
hepatocellular carcinoma; mCRPC, metastatic castration-resistant prostate cancer; NSCLC, non–small cell lung cancer; Panc, pancreatic cancer; PD,
progressive disease; PR, partial response; RCC, renal cell carcinoma; SD, stable disease.
3 additional patients. Of the 10 patients who had SD, 5 (50%) were on treatment −31% but also a new lesion in a mixed response (Fig. 1D). One additional PR
>20 weeks. Among 5 patients with CCA, 3 had at least two assessments of SD, was achieved in a patient with heavily pretreated CCA that was PD-L1 nega-
including 1 patient who was on treatment for almost 10 months before discon- tive, mismatch repair proficient, with a tumor mutational burden of 10 mut/Mb;
tinuing due to symptomatic deterioration while still meeting RECIST for SD he had received six prior lines of systemic therapy before study enrollment
(Fig. 1A; Supplementary Fig. S2). (Fig. 2B).
Of the 15 response-evaluable patients in the combination therapy cohort, 3 Notably, both patients with RCC who responded to the TPST-1120 combina-
patients (20.0%) achieved a PR (1 confirmed), while a best response of SD oc- tion regimen (and the patient with RCC with a −31% mixed response) were
curred in 3 patients (20%), and 9 patients (60%) had PD. All responders were previously treated with at least one anti-PD-1 containing regimen with a best
treated at the two highest doses of TPST-1120 (≥400 mg twice daily) for an ORR response of SD and discontinued the most recent anti-PD-1 therapy for dis-
of 30% (3/10) at the 400 and 600 mg doses. In addition, of 4 evaluable patients ease progression. The RCC responder who achieved a −54% RECIST PR at
with RCC, 2 achieved a PR for an ORR of 50% in this indication (Fig. 2A for the first tumor assessment (8 weeks of treatment) had previously received
one of the responders) and a third demonstrated a target lesion regression of first-line ipilimumab + nivolumab followed by cabozantinib and everolimus,
1104 Cancer Res Commun; 4(4) April 2024 https://doi.org/10.1158/2767-9764.CRC-24-0082 | CANCER RESEARCH COMMUNICATIONS
PPARα Inhibition in Patients with Advanced Solid Cancers
discontinuing each regimen for PD after a best response of SD. The RCC re- (FDR P < 0.05, effect size > 0.5, Fig. 3A). Similar associations between expres-
sponder who achieved a −30% RECIST PR had previously received first-line sion levels of these genes and TPST-1120 exposure were also observed on cycle 3
pembrolizumab + axitinib followed by cabozantinib and discontinued both day 1 (Supplementary Fig. S3). Modulated genes included FCGRIIA (CD32; ref.
regimens for disease progression after a best response of SD. 20), ITGAX (CD11c; ref. 21), TAP (22, 23), and TNFRSFA (CD120a; ref. 24), all
of which are gene targets of transcription factors inhibited by PPARα through
Biomarker Exploration transrepression (Table 3). Analysis of gene expression changes by exposure ter-
Analysis of differential expression levels of 780 genes in whole blood specimens tile demonstrated that median expression levels of three of the four target genes
revealed that four were increased as a function of TPST-1120 AUC0–24 on day 8 at the middle exposure tertile, 11,818–20,749 nghour/mL, were statistically
elevated above that observed in the lowest tertile (P < 0.05 by Wilcoxon pair- Lipid analysis revealed on-treatment elevation in circulating free fatty acids
wise comparison, Fig. 3B). This corresponds to 60% of patients with TPST-1120 (FFA) from baseline to day 57 and day 85 in patients demonstrating PRs that
steady-state exposures of at least 11,819 nghour/mL who demonstrated target were not detected in patients with a best response of PD/SD (Supplementary
gene elevations above baseline levels and identifies a minimum exposure for Fig. S5).
induction of pharmacodynamic activity.
LDA of day 8 gene expression levels among BOR categories revealed clear dis- OBD Selection
tinctions in expression patterns between PD or SD patients and PR patients The 600 mg twice daily dose of TPST-1120 was determined as the OBD for both
(Supplementary Fig. S4A). PR patients were observed to express statisti- monotherapy and combination therapy regimens. Analysis of pharmacokinet-
cally decreased levels of CFB and PVR (CD155) and increased levels of ics across doses and for both monotherapy and combination showed a linear
APOE, MAGEA, RORC, and SYT (P < 0.05 by Mann–Whitney U test; relationship between TPST-1120 dose and plasma exposure, with no saturation
Supplementary Table S3; Supplementary Fig. S4B). at 600 mg twice daily, the highest dose level tested. Pharmacodynamic analysis
FCGR2A Fc-γ RIIa, CD32 Enhances antibody-dependent cytotoxicity of tumor 0.047, 0.732 STAT1 (20)
cells, increased phagocytosis and cytokine release
by myeloid cells (25)
ITGAX Integrin α-X, CD11c Marker of conventional dendritic cells that 0.049, 0.566 Fos, Jun, C/EBP (21)
cross-present tumor antigens, enhances
phagocytosis of tumor cells by macrophages (26)
TAP1 Transporter associated with Increases endogenous antigen processing and 0.049, 0.550 STAT1 (22), NFκB (23)
antigen processing-1 presentation by MHC-I molecules
TNFRSF1A TNFα R1, CD120a Enhances responsiveness to TNFα, activates 0.047, 0.648 STAT3 (24)
NFκB-responsive genes (27)
1106 Cancer Res Commun; 4(4) April 2024 https://doi.org/10.1158/2767-9764.CRC-24-0082 | CANCER RESEARCH COMMUNICATIONS
PPARα Inhibition in Patients with Advanced Solid Cancers
of the association between TPST-1120 steady-state exposures and gene expres- factors that are subject to PPARα transrepressive activities, including NFκB
sion changes in peripheral blood demonstrated exposure-dependent increases and STAT1. This transcriptional dose–response allows for establishment of a
in expression of a subset of genes known to be regulated by transcription factors minimum exposure threshold for pharmacodynamic activity corresponding
transrepressed by PPARα, and the identified minimum exposure for induction to a TPST-1120 dose of at least 400 mg twice daily. Consistent with the ex-
of pharmacodynamic activity was achieved in all patients receiving TPST-1120 posure threshold, all patients who achieved a PR were dosed with TPST-1120
at 400 or 600 mg twice daily (Supplementary Fig. S1). Review of safety data at ≥400 mg twice daily. Further exploratory analyses of associations between
showed no evidence of dose-dependent toxicity through the highest dose tested BOR and changes in both gene expression and lipids in study patients showed
of 600 mg twice daily, either for single agent TPST-1120 or when administered increases in circulating FFA levels, indicating a reduction in lipid catabolism,
with nivolumab. Finally, although limited by small numbers, RECIST responses and increases in RORC, encoding RORγt, the master transcriptional regulator
in the combination cohort all occurred at the two highest TPST-1120 dose levels of Th17 cells, consistent with literature reports that Th17 cells are increased in
tested, consistent with dose-responsive antitumor activity. PPARα-deficient animals (35, 36).
Authors’ Disclosures Next Cure, Silverback, and Takeda outside the submitted work; and consult-
ing for AstraZeneca, Bioline Rx, DNAMx, Genentech, Roche, GPCR, Gilead,
M. Yarchoan reports grants and personal fees from Genentech; grants from Immune Onc, Immunitas, Immutep, Lilly, Macrogenics Mersana, Shionogi
Bristol Myers Squibb and Incyte; personal fees from Exelixis, Eisai, As- member SAB Bioline RX member CAB Immutep potential for equity with
traZeneca, Replimune, Hepion, Lantheus; and other from Adventris outside Ankyra Therapeutics potential for future royalties from Molecuvax. R. Stagg
the submitted work. J.D. Powderly reports personal fees and other from reports personal fees and other from Tempest outside the submitted work;
Tempest Therapeutics during the conduct of the study; other from Carolina and Clinical consultant for Tempest Therapeutics. S. Smith reports personal
BioOncology Institute, PLLC, BioCytics Inc., Genentech Roche, AstraZeneca fees from Tempest Therapeutics during the conduct of the study. C.C. Whiting
Medimmune, AbbVie, Aavocyte, AavoBioCytics, Phanes Therapeutics, BMS, reports other from Tempest Therapeutics outside the submitted work; in addi-
EMD Serono, Macrogenics, Top Alliance BioSciences, Alkermes, Calico, tion, C.C. Whiting has a patent to us2023/078674 pending. A. Moon reports
Atreca, Precision for Medicine, Stem Cell, Sequenom, Replimuune, Engi- personal fees, non-financial support, and other from Tempest Therapeutics
neered BioPharmaceuticals, Therapeutic Brainpower, Merck, PIOMA, Xilis, during the conduct of the study. P. Prasit reports other from Tempest Therapeu-
Repertoire Immune Medicines, NextCure, Xilio Therapeutics, Nuvation Bio, tics outside the submitted work. N. Standifer reports other from AstraZeneca,
1108 Cancer Res Commun; 4(4) April 2024 https://doi.org/10.1158/2767-9764.CRC-24-0082 | CANCER RESEARCH COMMUNICATIONS
PPARα Inhibition in Patients with Advanced Solid Cancers
References
1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 19. Su B, Bettcher LF, Hsieh W-Y, Hornburg D, Pearson MJ, Blomberg N,
2011;144: 646-74. et al. A DMS shotgun lipidomics workflow application to facilitate high-
throughput, comprehensive lipidomics. J Am Soc Mass Spectrom 2021;32:
34. Ravi P, Mantia C, Su C, Sorenson K, Elhag D, Rathi N, et al. Evaluation of the 39. Devchand PR, Keller H, Peters JM, Vazquez M, Gonzalez FJ, Wahli W. The
safety and efficacy of immunotherapy rechallenge in patients with renal cell PPARα–leukotriene B4 pathway to inflammation control. Nature 1996;384:
carcinoma. JAMA Oncol 2020;6: 1606-10. 39-43.
35. Chang H, Zhao F, Xie X, Liao Y, Song Y, Liu C, et al. PPARα suppresses 40. Cuzzocrea S, Mazzon E, Paola RD, Peli A, Bonato A, Britti D, et al. The role of
Th17 cell differentiation through IL-6/STAT3/RORγt pathway in experimental the peroxisome proliferator-activated receptor-α (PPAR-α) in the regulation of
autoimmune myocarditis. Exp Cell Res 2019;375: 22-30. acute inflammation. J Leukoc Biol 2006;79: 999-1010.
36. Wei P, Kou W, Fu J, Chen Z, Pan F. Pparα knockout in mice increases the 41. Gervois P, Mansouri RM. PPARα as a therapeutic target in inflammation-
Th17 development by facilitating the IKKα/RORγt and IKKα/Foxp3 complexes. associated diseases. Expert Opin Ther Targets 2012;16: 1113-25.
Commun Biol 2023;6: 721. 42. Pawlak M, Baugé E, Bourguet W, Bosscher K, Lalloyer F, Tailleux A, et al. The
37. Xie G, Song Y, Li N, Zhang Z, Wang X, Liu Y, et al. Myeloid peroxisome transrepressive activity of peroxisome proliferator-activated receptor alpha is
proliferator-activated receptor α deficiency accelerates liver regeneration via necessary and sufficient to prevent liver fibrosis in mice. Hepatology 2014;60:
IL-6/STAT3 pathway after 2/3 partial hepatectomy in mice. Hepatobiliary Surg 1593-606.
Nutr 2022;11: 199-211. 43. Tempest Therapeutics Inc. Tempest releases new data demonstrating su-
38. Zhang MA, Ahn JJ, Zhao FL, Selvanantham T, Mallevaey T, Stock N, et al. periority of TPST-1120 arm across multiple study endpoints in randomized
Antagonizing peroxisome proliferator–activated receptor α activity selec- first-line HCC study; 2023. Available from: https://ir.tempesttx.com/news-
tively enhances Th1 immunity in male mice. J Immunol 2015;195: 5189- releases/news-release-details/tempest-releases-new-data-demonstrating-
1110 Cancer Res Commun; 4(4) April 2024 https://doi.org/10.1158/2767-9764.CRC-24-0082 | CANCER RESEARCH COMMUNICATIONS