1
|
Garrido-Castro AC, Graham N, Ali LR, Herold C, Desrosiers J, Do K, Parsons H, Li T, Goel S, DiLullo M, Wrabel E, Williams AJ, Liu JF, Mittendorf EA, Dougan SK, Tayob N, Shapiro GI, Tolaney SM. Phase I study of ribociclib (CDK4/6 inhibitor) with spartalizumab (PD-1 inhibitor) with and without fulvestrant in metastatic hormone receptor-positive breast cancer or advanced ovarian cancer. J Immunother Cancer 2025; 13:e010430. [PMID: 40010764 PMCID: PMC11865738 DOI: 10.1136/jitc-2024-010430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Preclinical evidence suggests that cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors enhance antitumor immunity. We conducted a phase I trial of ribociclib (CDK4/6 inhibitor) plus spartalizumab (PD-1 inhibitor) in patients with hormone receptor (HR)-positive/HER2-negative metastatic breast cancer (MBC) or advanced ovarian cancer (AOC). The combination was also evaluated with fulvestrant in MBC. METHODS In Cohort A, ribociclib was administered on Days 1-21 (28-day cycle) starting at 400 mg, and spartalizumab at 400 mg on Day 1. Dose escalation was followed by expansion in AOC. Fulvestrant was added (Cohort B) with a safety run-in followed by expansion in MBC. Primary objectives were to determine the maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D), and safety and tolerability of the combinations. RESULTS 33 patients enrolled (n=18, Cohort A; n=15, Cohort B). The RP2D of ribociclib in both cohorts was 600 mg. Treatment-related adverse events in >20% of patients in either cohort were neutropenia, fatigue, anemia, thrombocytopenia, hypertransaminasemia, maculopapular rash, fatigue, and nausea. Hypertransaminasemia occurred in 66.7% (AST) and 46.7% (ALT) of patients in Cohort B, including 46.7% and 40.0%, respectively, of grade 3 or 4 events. Two confirmed partial responses were observed (13.3%) in Cohort B, in patients with low baseline serum thymidine kinase activity, coupled with an increase on-treatment. Peripheral blood flow cytometry across patients demonstrated on-target drug binding with increases in PD-1 occupancy and activated CD8+ T cells during treatment, irrespective of response. PD-L1-positivity, tumor-infiltrating lymphocytes, or tumor mutational burden did not correlate with progression-free survival (PFS). Several copy-number variations detected with next-generation sequencing correlated with PFS. CONCLUSIONS Ribociclib with spartalizumab and fulvestrant showed limited efficacy and elevated hepatotoxicity, precluding further development. Correlative analyses revealed treatment-induced immunological effects, and genomic alterations associated with PFS.
Collapse
Affiliation(s)
- Ana C Garrido-Castro
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Noah Graham
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Lestat R Ali
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Christina Herold
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Takeda Oncology, Cambridge, Massachusetts, USA
| | - Jennifer Desrosiers
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Khanh Do
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Heather Parsons
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Tianyu Li
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Shom Goel
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Molly DiLullo
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
| | - Eileen Wrabel
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
| | - Amy J Williams
- Biovica International AB, Uppsala, Uppsala County, Sweden
| | - Joyce F Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Stephanie K Dougan
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nabihah Tayob
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Luo T, Fan Z, Zeng A, Wang A, Pan Y, Xu Y, Chen H, Chen W, Nie D, Lin J, Huang A, Gong M, Huang Y, Ding Y, Zhu X, Rao L, Wang J. Biomimetic Targeted Co-Delivery System Engineered from Genomic Insights for Precision Treatment of Osteosarcoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410427. [PMID: 39555699 PMCID: PMC11727257 DOI: 10.1002/advs.202410427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/28/2024] [Indexed: 11/19/2024]
Abstract
The high heterogeneity and severe side effects of chemotherapy are major factors contributing to the failure of osteosarcoma treatment. Herein, a comprehensive genomic analysis is conducted, and identified two prominent characteristics of osteosarcoma: significant cyclin-dependent kinases 4 (CDK4) amplification and homologous recombination repair deficiency. Based on these findings, a co-delivery system loaded with CDK4/6 inhibitors and poly ADP-ribose polymerase (PARP) inhibitors is designed. By employing metal-organic frameworks (MOFs) as carriers, issue of drug insolubility is effectively addressed, while also enabling controlled release in response to the tumor microenvironment. To enhance targeting capability and biocompatibility, the MOFs are further coated with a bio-membrane targeting B7H3. This targeted biomimetic co-delivery system possesses several key features: 1) it can precisely target osteosarcoma with high B7H3 expression; 2) the combination of CDK4/6 inhibitors and PARP inhibitors exhibits synergistic effects, significantly impairing tumor's DNA repair capacity; and 3) the system has the potential for combination with photodynamic therapy, amplifying DNA repair defects to maximize tumor cell eradication. Furthermore, it is observed that this co-delivery system can activate immune microenvironment, increasing CD8+ T cell infiltration and converting osteosarcoma from an immune-cold to an immune-hot tumor. In summary, the co-delivery system is an effective therapeutic strategy and holds promise as a novel approach for osteosarcoma treatment.
Collapse
Affiliation(s)
- Tianqi Luo
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Zhijin Fan
- Institute for Engineering MedicineKunming Medical UniversityKunming650500China
| | - Anyu Zeng
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Anqi Wang
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Yuanwei Pan
- Institute of Chemical BiologyShenzhen Bay LaboratoryShenzhen518132China
| | - Yanyang Xu
- Department of Joint SurgeryGuangzhou First People's HospitalSchool of MedicineSouth China University of TechnologyGuangzhou510060China
| | - Hongmin Chen
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Weiqing Chen
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Dingmeng Nie
- Institute of Chemical BiologyShenzhen Bay LaboratoryShenzhen518132China
| | - Jiaming Lin
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Anfei Huang
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Ming Gong
- Department of Pediatric OrthopaedicsGuangzhou Women and Children's Medical CenterGuangdong Provincial Clinical Research Center for Child HealthGuangzhou Medical UniversityGuangzhou510630China
| | - Yufeng Huang
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Yun Ding
- Department of Breast OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Xiaojun Zhu
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Lang Rao
- Institute of Chemical BiologyShenzhen Bay LaboratoryShenzhen518132China
| | - Jin Wang
- Department of Musculoskeletal OncologyState Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060China
| |
Collapse
|
3
|
Wang Z, Liu Y, Wang K, Ma L. Efficacy and safety of PD-1 and PD-L1 inhibitors in advanced colorectal cancer: a meta-analysis of randomized controlled trials. BMC Gastroenterol 2024; 24:461. [PMID: 39696009 DOI: 10.1186/s12876-024-03554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND PD-1 and PD-L1 inhibitors have emerged as promising therapies for advanced colorectal cancer (CRC), but their efficacy and safety profiles require further evaluation. This meta-analysis aims to assess the efficacy and safety of PD-1/PD-L1 inhibitors in this patient population. METHODS A systematic review and meta-analysis were conducted following PRISMA guidelines, with data sourced from PubMed, Embase, CENTRAL, Web of Science, and CNKI up to August 3, 2024. Nine randomized controlled trials (RCTs) involving 1680 patients were included. The primary outcomes were overall survival (OS), progression-free survival (PFS) and objective response rate (ORR), while safety was assessed through adverse events (AEs) and grade ≥ 3 AEs. Effect sizes were calculated using mean differences (MD) and risk ratios (RR) with 95% confidence intervals (CIs). RESULTS Overall, the meta-analysis showed that PD-1/PD-L1 inhibitors did not significantly extend OS (MD = 0.86, 95% CI: -0.55, 2.27), but they significantly improved PFS (MD = 2.53, 95% CI: 0.92, 4.15). Additionally, PD-1/PD-L1 inhibitors did not significantly increase the ORR compared to controls (RR = 1.19, 95% CI: 0.99, 1.44). In terms of safety, PD-1/PD-L1 inhibitors did not significantly increase the incidence of overall AEs. Subgroup analysis further indicated that PD-1 inhibitors significantly improved OS (MD = 1.24, 95% CI: 0.20, 2.29) and PFS (MD = 6.27, 95% CI: 0.56, 11.97), while PD-L1 inhibitors did not have a significant impact on these outcomes. Additionally, PD-L1 inhibitors were associated with a higher risk of grade ≥ 3 AEs (RR = 1.29, 95% CI: 1.07, 1.57), a risk not observed with PD-1 inhibitors. CONCLUSION PD-1 inhibitors significantly improve PFS and OS in advanced CRC, making them a preferable option over PD-L1 inhibitors, which show limited efficacy and a higher risk of severe AEs. These findings support prioritizing PD-1 inhibitors in clinical practice for this patient group, while caution is warranted with PD-L1 inhibitors due to their safety concerns. TRIAL REGISTRATION PROSPERO (CRD42024611696).
Collapse
Affiliation(s)
- Zhenzi Wang
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yuan Liu
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Kedi Wang
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Liyan Ma
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
4
|
Oberstein PE, Dias Costa A, Kawaler EA, Cardot-Ruffino V, Rahma OE, Beri N, Singh H, Abrams TA, Biller LH, Cleary JM, Enzinger P, Huffman BM, McCleary NJ, Perez KJ, Rubinson DA, Schlechter BL, Surana R, Yurgelun MB, Wang SJ, Remland J, Brais LK, Bollenrucher N, Chang E, Ali LR, Lenehan PJ, Dolgalev I, Werba G, Lima C, Keheler CE, Sullivan KM, Dougan M, Hajdu C, Dajee M, Pelletier MR, Nazeer S, Squires M, Bar-Sagi D, Wolpin BM, Nowak JA, Simeone DM, Dougan SK. Blockade of IL1β and PD1 with Combination Chemotherapy Reduces Systemic Myeloid Suppression in Metastatic Pancreatic Cancer with Heterogeneous Effects in the Tumor. Cancer Immunol Res 2024; 12:1221-1235. [PMID: 38990554 PMCID: PMC11369625 DOI: 10.1158/2326-6066.cir-23-1073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/15/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024]
Abstract
Innate inflammation promotes tumor development, although the role of innate inflammatory cytokines in established human tumors is unclear. Herein, we report clinical and translational results from a phase Ib trial testing whether IL1β blockade in human pancreatic cancer would alleviate myeloid immunosuppression and reveal antitumor T-cell responses to PD1 blockade. Patients with treatment-naïve advanced pancreatic ductal adenocarcinoma (n = 10) were treated with canakinumab, a high-affinity monoclonal human antiinterleukin-1β (IL1β), the PD1 blocking antibody spartalizumab, and gemcitabine/n(ab)paclitaxel. Analysis of paired peripheral blood from patients in the trial versus patients receiving multiagent chemotherapy showed a modest increase in HLA-DR+CD38+ activated CD8+ T cells and a decrease in circulating monocytic myeloid-derived suppressor cells (MDSC) by flow cytometry for patients in the trial but not in controls. Similarly, we used patient serum to differentiate monocytic MDSCs in vitro and showed that functional inhibition of T-cell proliferation was reduced when using on-treatment serum samples from patients in the trial but not when using serum from patients treated with chemotherapy alone. Within the tumor, we observed few changes in suppressive myeloid-cell populations or activated T cells as assessed by single-cell transcriptional profiling or multiplex immunofluorescence, although increases in CD8+ T cells suggest that improvements in the tumor immune microenvironment might be revealed by a larger study. Overall, the data indicate that exposure to PD1 and IL1β blockade induced a modest reactivation of peripheral CD8+ T cells and decreased circulating monocytic MDSCs; however, these changes did not lead to similarly uniform alterations in the tumor microenvironment.
Collapse
Affiliation(s)
- Paul E. Oberstein
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Emily A. Kawaler
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Surgery, NYU Langone Health, New York, New York.
| | - Victoire Cardot-Ruffino
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
| | - Osama E. Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Nina Beri
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Harshabad Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Thomas A. Abrams
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Leah H. Biller
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Peter Enzinger
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Brandon M. Huffman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Nadine J. McCleary
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Kimberly J. Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Douglas A. Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Benjamin L. Schlechter
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Rishi Surana
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Matthew B. Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - S. Jennifer Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Joshua Remland
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Lauren K. Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Naima Bollenrucher
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Eugena Chang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Lestat R. Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Patrick J. Lenehan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
| | - Igor Dolgalev
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Gregor Werba
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Surgery, NYU Langone Health, New York, New York.
| | - Cibelle Lima
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - C. Elizabeth Keheler
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Keri M. Sullivan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Michael Dougan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Cristina Hajdu
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Pathology, NYU Langone Health, New York, New York.
| | - Maya Dajee
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts.
| | - Marc R. Pelletier
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts.
| | | | | | - Dafna Bar-Sagi
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Jonathan A. Nowak
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Diane M. Simeone
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Surgery, NYU Langone Health, New York, New York.
| | - Stephanie K. Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
5
|
Kim NH, Lee J, Kim SH, Kang SH, Bae S, Yu CH, Seo J, Kim HT. ALK5/VEGFR2 dual inhibitor TU2218 alone or in combination with immune checkpoint inhibitors enhances immune-mediated antitumor effects. Cancer Immunol Immunother 2024; 73:190. [PMID: 39105882 PMCID: PMC11303640 DOI: 10.1007/s00262-024-03777-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024]
Abstract
Transforming growth factor β (TGFβ) is present in blood of patients who do not respond to anti-programmed cell death (ligand) 1 [PD-(L)1] treatment, and through synergy with vascular endothelial growth factor (VEGF), it helps to create an environment that promotes tumor immune evasion and immune tolerance. Therefore, simultaneous inhibition of TGFβ/VEGF is more effective than targeting TGFβ alone. In this study, the dual inhibitory mechanism of TU2218 was identified through in vitro analysis mimicking the tumor microenvironment, and its antitumor effects were analyzed using mouse syngeneic tumor models. TU2218 directly restored the activity of damaged cytotoxic T lymphocytes (CTLs) and natural killer cells inhibited by TGFβ and suppressed the activity and viability of regulatory T cells. The inactivation of endothelial cells induced by VEGF stimulation was completely ameliorated by TU2218, an effect not observed with vactosertib, which inhibits only TGFβ signaling. The combination of TU2218 and anti-PD1 therapy had a significantly greater antitumor effect than either drug alone in the poorly immunogenic B16F10 syngeneic tumor model. The mechanism of tumor reduction was confirmed by flow cytometry, which showed upregulated VCAM-1 expression in vascular cells and increased influx of CD8 + CTLs into the tumor. As another strategy, combination of anti-CTLA4 therapy and TU2218 resulted in high complete regression (CR) rates in CT26 and WEHI-164 tumor models. In particular, immunological memory generated by the combination of anti-CTLA4 and TU2218 in the CT26 model prevented the development of tumors after additional tumor cell transplantation, suggesting that the TU2218-based combination has therapeutic potential in immunotherapy.
Collapse
Affiliation(s)
- Nam-Hoon Kim
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jihyun Lee
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seung-Hyun Kim
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seong-Ho Kang
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Sowon Bae
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Chan-Hee Yu
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jeongmin Seo
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hun-Taek Kim
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
6
|
Preite NW, Borges BM, Kaminski VDL, Ayupe MC, Gonçalves LM, dos Santos BV, Fonseca DLM, Filgueiras IS, Salgado CL, Muxel SM, Cabral-Marques O, da Fonseca DM, Loures FV, Calich VLG. Blocking the CTLA-4 and PD-1 pathways during pulmonary paracoccidioidomycosis improves immunity, reduces disease severity, and increases the survival of infected mice. Front Immunol 2024; 15:1347318. [PMID: 38500881 PMCID: PMC10945025 DOI: 10.3389/fimmu.2024.1347318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/14/2024] [Indexed: 03/20/2024] Open
Abstract
Immune checkpoint pathways, i.e., coinhibitory pathways expressed as feedback following immune activation, are crucial for controlling an excessive immune response. Cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death protein-1 (PD-1) are the central classical checkpoint inhibitory (CPI) molecules used for the control of neoplasms and some infectious diseases, including some fungal infections. As the immunosuppression of severe paracoccidioidomycosis (PCM), a chronic granulomatous fungal disease, was shown to be associated with the expression of coinhibitory molecules, we hypothesized that the inhibition of CTLA-4 and PD-1 could have a beneficial effect on pulmonary PCM. To this end, C57BL/6 mice were infected with Paracoccidioides brasiliensis yeasts and treated with monoclonal antibodies (mAbs) α-CTLA-4, α-PD-1, control IgG, or PBS. We verified that blockade of CTLA-4 and PD-1 reduced the fungal load in the lungs and fungal dissemination to the liver and spleen and decreased the size of pulmonary lesions, resulting in increased survival of mice. Compared with PBS-treated infected mice, significantly increased levels of many pro- and anti-inflammatory cytokines were observed in the lungs of α-CTLA-4-treated mice, but a drastic reduction in the liver was observed following PD-1 blockade. In the lungs of α-CPI and IgG-treated mice, there were no changes in the frequency of inflammatory leukocytes, but a significant reduction in the total number of these cells was observed. Compared with PBS-treated controls, α-CPI- and IgG-treated mice exhibited reduced pulmonary infiltration of several myeloid cell subpopulations and decreased expression of costimulatory molecules. In addition, a decreased number of CD4+ and CD8+ T cells but sustained numbers of Th1, Th2, and Th17 T cells were detected. An expressive reduction in several Treg subpopulations and their maturation and suppressive molecules, in addition to reduced numbers of Treg, TCD4+, and TCD8+ cells expressing costimulatory and coinhibitory molecules of immunity, were also detected. The novel cellular and humoral profiles established in the lungs of α-CTLA-4 and α-PD-1-treated mice but not in control IgG-treated mice were more efficient at controlling fungal growth and dissemination without causing increased tissue pathology due to excessive inflammation. This is the first study demonstrating the efficacy of CPI blockade in the treatment of pulmonary PCM, and further studies combining the use of immunotherapy with antifungal drugs are encouraged.
Collapse
Affiliation(s)
| | | | | | - Marina Caçador Ayupe
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Leonardo Mandu Gonçalves
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | | | | | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Caio Loureiro Salgado
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Sandra Marcia Muxel
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine (USP), São Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, Brazil
| | - Denise Morais da Fonseca
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Flávio Vieira Loures
- Institute of Science and Technology, Federal University of São Paulo, São Paulo, Brazil
| | - Vera Lúcia Garcia Calich
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
7
|
Ali LR, Lenehan PJ, Cardot-Ruffino V, Dias Costa A, Katz MH, Bauer TW, Nowak JA, Wolpin BM, Abrams TA, Patel A, Clancy TE, Wang J, Mancias JD, Reilley MJ, Stucky CCH, Bekaii-Saab TS, Elias R, Merchant N, Slingluff CL, Rahma OE, Dougan SK. PD-1 Blockade Induces Reactivation of Nonproductive T-Cell Responses Characterized by NF-κB Signaling in Patients with Pancreatic Cancer. Clin Cancer Res 2024; 30:542-553. [PMID: 37733830 PMCID: PMC10831338 DOI: 10.1158/1078-0432.ccr-23-1444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/28/2023] [Accepted: 09/19/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) trials have evaluated CTLA-4 and/or PD-(L)1 blockade in patients with advanced disease in which bulky tumor burden and limited time to develop antitumor T cells may have contributed to poor clinical efficacy. Here, we evaluated peripheral blood and tumor T cells from patients with PDAC receiving neoadjuvant chemoradiation plus anti-PD-1 (pembrolizumab) versus chemoradiation alone. We analyzed whether PD-1 blockade successfully reactivated T cells in the blood and/or tumor to determine whether lack of clinical benefit could be explained by lack of reactivated T cells versus other factors. EXPERIMENTAL DESIGN We used single-cell transcriptional profiling and TCR clonotype tracking to identify TCR clonotypes from blood that match clonotypes in the tumor. RESULTS PD-1 blockade increases the flux of TCR clonotypes entering cell cycle and induces an IFNγ signature like that seen in patients with other GI malignancies who respond to PD-1 blockade. However, these reactivated T cells have a robust signature of NF-κB signaling not seen in cases of PD-1 antibody response. Among paired samples between blood and tumor, several of the newly cycling clonotypes matched activated T-cell clonotypes observed in the tumor. CONCLUSIONS Cytotoxic T cells in the blood of patients with PDAC remain sensitive to reinvigoration by PD-1 blockade, and some have tumor-recognizing potential. Although these T cells proliferate and have a signature of IFN exposure, they also upregulate NF-κB signaling, which potentially counteracts the beneficial effects of anti-PD-1 reinvigoration and marks these T cells as non-productive contributors to antitumor immunity. See related commentary by Lander and DeNardo, p. 474.
Collapse
Affiliation(s)
- Lestat R. Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Patrick J. Lenehan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Victoire Cardot-Ruffino
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Matthew H.G. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Todd W. Bauer
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Jonathan A. Nowak
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Thomas A. Abrams
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Anuj Patel
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Thomas E. Clancy
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jiping Wang
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joseph D. Mancias
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Matthew J. Reilley
- Division of Hematology and Oncology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | | | | | - Rawad Elias
- Hartford Healthcare Cancer Institute, Hartford, Connecticut
| | - Nipun Merchant
- Department of Surgery, University of Miami, Miami, Florida
| | - Craig L. Slingluff
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Osama E. Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Stephanie K. Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
8
|
Wang Y, Zhu J, Zhou N, Wang Y, Zhang X. Changes in T lymphocyte subsets predict the efficacy of atezolizumab in advanced non-small cell lung cancer: a retrospective study. J Thorac Dis 2023; 15:5669-5679. [PMID: 37969284 PMCID: PMC10636460 DOI: 10.21037/jtd-23-1169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/01/2023] [Indexed: 11/17/2023]
Abstract
Background It has remained unclear how programmed cell death ligand 1 (PD-L1) inhibitors affect peripheral blood lymphocyte (PBL) subsets in patients with advanced non-small cell lung cancer (NSCLC). This study assessed the predictive and prognostic value of PBL subsets in patients with advanced NSCLC who were treated with atezolizumab. Methods A total of 30 patients with advanced NSCLC treated with atezolizumab were selected as the observation group, and 30 healthy individuals were chosen as the control group during same period. Flow cytometry was used to detect lymphocyte subsets before and after treatment. The relationship between the changes of lymphocyte subsets and atezolizumab in the treatment of NSCLC was analyzed and calculated. Results Before treatment, compared with the control group, the number of CD3+, CD4+ T, and CD4+/CD8+ indexes in the observation group were significantly decreased, whereas the level of CD8+ was significantly increased. The number of CD3+, CD4+ T, and CD4+/CD8+ indexes gradually increased with the process of atezolizumab treatment, whereas the number of CD8+ T gradually decreased. After the 4 cycles, the number of CD3+, CD4+ T, and CD4+/CD8+ indexes were significantly increased, and the number of CD8+ was significantly decreased. In the observation group, 22 patients achieved partial response (PR)/stable disease (SD) and 8 patients achieved progressive disease (PD) after 4 cycles of atezolizumab treatment. Before treatment, there were no significant differences in the level of lymphocyte subsets between those who achieved PR/SD or PD. However, a significant difference in the level of lymphocyte subsets appeared after 4 cycles of atezolizumab treatment. Among the 22 patients who achieved PR/SD, the number of CD3+, CD4+ T, and CD4+/CD8+ indexes were significantly increased, whereas the number of CD8+ T lymphocytes was significantly decreased. Meanwhile, the 8 patients who achieved PD displayed different results. In addition, ROC curve combined detection of CD3+, CD4+, and CD8+ T [area under the curve (AUC) =0.9018, P<0.0001] showed good predictive ability for the efficacy of atezolizumab in advanced NSCLC. Conclusions Atezolizumab may alter the level of lymphocyte subsets in patients with advanced NSCLC, and the changes in lymphocyte subsets may predict the efficacy of atezolizumab for these patients.
Collapse
Affiliation(s)
- Yuyang Wang
- Qingdao Medical College, Qingdao University, Qingdao, China
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjuan Zhu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Na Zhou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yun Wang
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaochun Zhang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Qian J, Zhang X, Wei B, Tang Z, Zhang B. The correlation between gut and intra-tumor microbiota and PDAC: Etiology, diagnostics and therapeutics. Biochim Biophys Acta Rev Cancer 2023; 1878:188943. [PMID: 37355177 DOI: 10.1016/j.bbcan.2023.188943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the lethal cancers in the world and its 5-year survival rate is <10%. Due to the unique TME and dense tissue structure, its curative efficacy is far from satisfactory,the immunotherapy is even more invalid. According to the recent studies, the gut and tumor microbiota have been proved to play a key role in the development, progression and prognosis of PDAC. Based on the differences of microbiome composition observed in PDAC patients and normal pancreas, many researches have been made focusing on the latent communication between gut and intra-tumor microbiota and PDAC. In this review, we will demonstrate the potential mechanism of the oncogenic effects of GM and IM and their crucial effects on modulating the TME. Besides, we focus on their interaction with chemotherapeutic and immunotherapeutic drugs and inducing the drug resistance, thus enlightening the promising role to be used to monitor the occurrence of PDAC, accurately modulate the immune environment to promote the therapeutic efficacy and predict the prognosis.
Collapse
Affiliation(s)
- Jiwei Qian
- The Fourth affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xin Zhang
- The Fourth affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Butian Wei
- The Fourth affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Zhe Tang
- The Fourth affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Bo Zhang
- The Second affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 31000, China.
| |
Collapse
|