1
|
Hecht JR, Papadopoulos KP, Falchook GS, Patel MR, Infante JR, Aljumaily R, Wong DJ, Autio KA, Wainberg ZA, Bauer TM, Javle M, Pant S, Bendell J, Hung A, Ratti N, VanVlasselaer P, Verma R, Leveque J, Rao S, Oft M, Naing A. Immunologic and tumor responses of pegilodecakin with 5-FU/LV and oxaliplatin (FOLFOX) in pancreatic ductal adenocarcinoma (PDAC). Invest New Drugs 2021; 39:182-192. [PMID: 32910338 PMCID: PMC8944136 DOI: 10.1007/s10637-020-01000-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/04/2020] [Indexed: 10/23/2022]
Abstract
Background Treatment options for pancreatic ductal adenocarcinoma (PDAC) are limited and checkpoint blockade inhibitors have been disappointing in this disease. Pegilodecakin has demonstrated single agent anti-tumor activity in immune-sensitive tumors. Phase 1 and preclinical data indicate synergy of pegilodecakin with 5-FU and platins. We assessed the safety and activity of pegilodecakin+FOLFOX in patients with PDAC. Methods IVY (NCT02009449) was an open-label phase 1b trial in the United States. Here we report on all enrolled patients from cohort C. Heavily pretreated patients were treated with pegilodecakin (self-administered subcutaneously daily at 2.5, 5, or 10 μg/kg) + 5-flurouracil/leucovorin/oxaliplatin (FOLFOX), dosed per manufacturers prescribing information, until tumor progression. Eligible patients had measurable disease per immune-related response criteria (irRC), were ≥ 18 years of age, and had ECOG performance status of 0 or 1. Patients were evaluated for primary(safety) and secondary (tumor response per irRC) endpoints. Results From 5 August 2014-12 July 2016, 39 patients enrolled in cohort C. All patients were evaluable for safety. In this advanced population, regimen had manageable toxicities with no immune-related adverse events (irAEs) greater than grade 1. The most common grade 3/4/5 TEAEs were thrombocytopenia (21[53.8%] of 39) and anemia (17[43.6%] of 39). In evaluable PDAC patients, the best overall response of pegilodecakin+FOLFOX was 3(14%) with CRs in 2(9%) patients. Conclusions Pegilodecakin+FOLFOX had an acceptable tolerability profile in PDAC, with no substantial irAEs seen, and promising efficacy with the combination yielding a 2-year OS of 24% (95% CI 10-42). These data led to the phase 3 study with pegilodecakin+FOLFOX as second-line therapy of PDAC (SEQUOIA).
Collapse
Affiliation(s)
- J Randolph Hecht
- David Geffen School of Medicine, UCLA, 10833 Le Conte Avenue, Santa Monica, CA, USA.
| | | | - Gerald S Falchook
- Sarah Cannon Research Institute at HealthONE, 1800 N Williams Street Suite 300, Denver, CO, USA
| | - Manish R Patel
- Sarah Cannon Research Institute/Florida Cancer Specialists, 600 N Cattlemen Road Suite 200, Sarasota, FL, USA
| | - Jeffrey R Infante
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, 300 20th Avenue N, Nashville, TN, USA
| | - Raid Aljumaily
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, 300 20th Avenue N, Nashville, TN, USA
- Stephenson Cancer Center of the University of Oklahoma, 800 NE 10th, Oklahoma City, OK, USA
| | - Deborah J Wong
- David Geffen School of Medicine, UCLA, 10833 Le Conte Avenue, Santa Monica, CA, USA
| | - Karen A Autio
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Zev A Wainberg
- David Geffen School of Medicine, UCLA, 10833 Le Conte Avenue, Santa Monica, CA, USA
| | - Todd M Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, 300 20th Avenue N, Nashville, TN, USA
| | - Milind Javle
- MD Anderson Cancer Center, 1515 Holcombe, Houston, TX, USA
| | - Shubham Pant
- Stephenson Cancer Center of the University of Oklahoma, 800 NE 10th, Oklahoma City, OK, USA
- MD Anderson Cancer Center, 1515 Holcombe, Houston, TX, USA
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, 300 20th Avenue N, Nashville, TN, USA
| | - Annie Hung
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Navneet Ratti
- ARMO BioSciences, a wholly owned subsidiary of Eli Lilly and Company, 575 Chesapeake Dr., Redwood City, CA, USA
| | - Peter VanVlasselaer
- ARMO BioSciences, a wholly owned subsidiary of Eli Lilly and Company, 575 Chesapeake Dr., Redwood City, CA, USA
| | - Rakesh Verma
- ARMO BioSciences, a wholly owned subsidiary of Eli Lilly and Company, 575 Chesapeake Dr., Redwood City, CA, USA
| | - Joseph Leveque
- David Geffen School of Medicine, UCLA, 10833 Le Conte Avenue, Santa Monica, CA, USA
| | - Sujata Rao
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Martin Oft
- ARMO BioSciences, a wholly owned subsidiary of Eli Lilly and Company, 575 Chesapeake Dr., Redwood City, CA, USA
| | - Aung Naing
- MD Anderson Cancer Center, 1515 Holcombe, Houston, TX, USA
| |
Collapse
|
2
|
Myeloid-Derived Suppressor Cells and Pancreatic Cancer: Implications in Novel Therapeutic Approaches. Cancers (Basel) 2019; 11:cancers11111627. [PMID: 31652904 PMCID: PMC6893814 DOI: 10.3390/cancers11111627] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/07/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a devastating human malignancy with poor prognosis and low survival rates. Several cellular mechanisms have been linked with pancreatic carcinogenesis and also implicated in inducing tumor resistance to known therapeutic regimens. Of various factors, immune evasion mechanisms play critical roles in tumor progression and impeding the efficacy of cancer therapies including PDAC. Among immunosuppressive cell types, myeloid-derived suppressor cells (MDSCs) have been extensively studied and demonstrated to not only support PDAC development but also hamper the anti-tumor immune responses elicited by therapeutic agents. Notably, recent efforts have been directed in devising novel approaches to target MDSCs to limit their effects. Multiple strategies including immune-based approaches have been explored either alone or in combination with therapeutic agents to target MDSCs in preclinical and clinical settings of PDAC. The current review highlights the roles and mechanisms of MDSCs as well as the implications of this immunomodulatory cell type as a potential target to improve the efficacy of therapeutic regimens for PDAC.
Collapse
|
3
|
Talbert EE, Cuitiño MC, Ladner KJ, Rajasekerea PV, Siebert M, Shakya R, Leone GW, Ostrowski MC, Paleo B, Weisleder N, Reiser PJ, Webb A, Timmers CD, Eiferman DS, Evans DC, Dillhoff ME, Schmidt CR, Guttridge DC. Modeling Human Cancer-induced Cachexia. Cell Rep 2019; 28:1612-1622.e4. [PMID: 31390573 PMCID: PMC6733019 DOI: 10.1016/j.celrep.2019.07.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/24/2019] [Accepted: 07/03/2019] [Indexed: 01/03/2023] Open
Abstract
Cachexia is a wasting syndrome characterized by pronounced skeletal muscle loss. In cancer, cachexia is associated with increased morbidity and mortality and decreased treatment tolerance. Although advances have been made in understanding the mechanisms of cachexia, translating these advances to the clinic has been challenging. One reason for this shortcoming may be the current animal models, which fail to fully recapitulate the etiology of human cancer-induced tissue wasting. Because pancreatic ductal adenocarcinoma (PDA) presents with a high incidence of cachexia, we engineered a mouse model of PDA that we named KPP. KPP mice, similar to PDA patients, progressively lose skeletal and adipose mass as a consequence of their tumors. In addition, KPP muscles exhibit a similar gene ontology as cachectic patients. We envision that the KPP model will be a useful resource for advancing our mechanistic understanding and ability to treat cancer cachexia.
Collapse
Affiliation(s)
- Erin E Talbert
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Maria C Cuitiño
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Katherine J Ladner
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
| | - Priyani V Rajasekerea
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
| | - Melissa Siebert
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
| | - Reena Shakya
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
| | - Gustavo W Leone
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael C Ostrowski
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brian Paleo
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Noah Weisleder
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Peter J Reiser
- Division of Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Amy Webb
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
| | - Cynthia D Timmers
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daniel S Eiferman
- Division of Trauma, Critical Care, and Burn, The Ohio State University, Columbus, OH 43210, USA
| | - David C Evans
- Division of Trauma, Critical Care, and Burn, The Ohio State University, Columbus, OH 43210, USA
| | - Mary E Dillhoff
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Division of Surgical Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Carl R Schmidt
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Division of Surgical Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Denis C Guttridge
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
4
|
Macarulla T, Blanc JF, Wang-Gillam A, Chen LT, Siveke JT, Mirakhur B, Chen J, de Jong FA. Liposomal irinotecan and 5-fluorouracil/leucovorin in older patients with metastatic pancreatic cancer - A subgroup analysis of the pivotal NAPOLI-1 trial. J Geriatr Oncol 2019; 10:427-435. [PMID: 30842038 DOI: 10.1016/j.jgo.2019.02.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/20/2018] [Accepted: 02/19/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Pancreatic cancer is a highly lethal disease predominantly affecting older patients. Characterization of outcomes in these patients may help optimise treatment decisions. The global, phase 3 NAPOLI-1 trial (NCT01494506) demonstrated an overall survival (OS) benefit with liposomal irinotecan and 5-flurouracil/leucovorin (nal-IRI + 5-FU/LV) versus 5-FU/LV. This subgroup analysis explored impact of age on outcomes in NAPOLI-1 patients, and nal-IRI + 5-FU/LV efficacy and safety in older patients. MATERIALS AND METHODS This exploratory, post-hoc analysis of the NAPOLI-1 trial included patients aged ≥eighteen years (no upper limit) with metastatic pancreatic adenocarcinoma that had progressed on gemcitabine-based therapy. Patients were stratified by age (cut-offs at 65, 70, and 75 years); OS and progression-free survival (PFS) were estimated by Kaplan-Meier analysis. RESULTS Of 417 randomized patients, 192 (46%), 110 (26%) and 43 (10%) were aged ≥65, ≥70 and ≥ 75 years, respectively. Mortality risk and risk of disease progression were similar in older and younger patients independent of treatment (HRs for median [m]OS/mPFS comparisons were 0.88/0.95 [<65 versus ≥65 years], 0.89/0.88 [<70 versus ≥70 years] and 1.04/0.98 [<75 versus ≥75 years]; P > .25). Reduced mortality/morbidity risk with nal-IRI + 5-FU/LV in older subgroups was in line with the wider population. No additional toxicities with nal-IRI + 5-FU/LV were observed in older patients: 86% of patients ≥75 years versus 69% <75 years required a dose delay or reduction due to toxicities (43% versus 32% dose reductions). DISCUSSION Results suggest that older patients with metastatic pancreatic adenocarcinoma that progressed on prior gemcitabine-based treatment can benefit from second-line therapy, supporting nal-IRI + 5-FU/LV treatment in older patients.
Collapse
Affiliation(s)
- Teresa Macarulla
- Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | | | | | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jens T Siveke
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; Cancer Consortium (DKTK, partner site Essen), German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Beloo Mirakhur
- Ipsen Biopharmaceuticals, Inc., Cambridge, MA, United States
| | - Jie Chen
- Shire plc, Cambridge, MA, United States
| | | |
Collapse
|
5
|
Cao R, Song W, Ye C, Liu X, Li L, Li Y, Yao H, Zhou X, Li L, Shao R. Internal enhancement of DNA damage by a novel bispecific antibody-drug conjugate-like therapeutics via blockage of mTOR and PD-L1 signal pathways in pancreatic cancer. Cancer Med 2019; 8:643-655. [PMID: 30681288 PMCID: PMC6382721 DOI: 10.1002/cam4.1974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/20/2018] [Accepted: 12/23/2018] [Indexed: 12/31/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a refractory malignant tumor with poor prognosis, limited chemotherapeutic efficacy, and only about 5% of 5-year survival rate. We generated a dual-targeting ligand-based lidamycin (DTLL) to investigate its efficacy against pancreatic cancer after preparing its precursor, DTLP. DTLP was shown specifically binding to EGFR and HER2 on cell surface, followed by endocytosis into cytoplasm of pancreatic cancer cells. DTLL significantly promoted apoptosis and cell cycle arrest at G2/M stages and inhibited cell proliferation. Pancreatic tumors of either MIA-paca-2 cell line-derived (CDX) or patient-derived xenograft (PDX) mouse models were significantly regressed in response to DTLL. It suggested that DTLL might be a highly potent bispecific antibody-drug conjugate (ADC)-like agent for pancreatic cancer therapy. LDM is known to function as an antitumor cytotoxic agent by its induction of DNA damage in cancer cells, therefore, DTLL, as its derivative, also showed similar cytotoxicity. However, we found that DTLL might reverse the AKT/mTOR feedback activation induced by LDM at the first time. The results from both in vitro and in vivo experiments suggested that DTLL enhanced DNA damage via EGFR/HER2-dependent blockage of PI3K/AKT/mTOR and PD-L1 signaling pathways in cancer cells, leading to the inhibition of cell proliferation and immunosurveillance escape from pancreatic tumor. Our studies on DTLL functional characterization revealed its novel mechanisms on internal enhancement of DNA damage and implied that DTLL might provide a promising targeted therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Rui Cao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Wenping Song
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Cheng Ye
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xiujun Liu
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Liang Li
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yi Li
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Hongjuan Yao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xiaofei Zhou
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Liang Li
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Rongguang Shao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| |
Collapse
|