1
|
Chumsri S, Larson JJ, Liu E, Tenner KS, Adams D, Weidner MT, Arnold AN, Haley DL, Advani P, Sideras K, Moreno-Aspitia A, Thompson EA, Perez EA, Knutson KL. Pembrolizumab in Combination with Binimetinib in Patients with Unresectable Locally Advanced or Metastatic Triple-Negative Breast Cancer. Clin Cancer Res 2025; 31:1885-1893. [PMID: 40053697 PMCID: PMC12079099 DOI: 10.1158/1078-0432.ccr-24-3001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/06/2025] [Accepted: 02/28/2025] [Indexed: 03/09/2025]
Abstract
PURPOSE Activation of the RAS/MAPK pathway is associated with reduced tumor-infiltrating lymphocytes and poor outcomes in triple-negative breast cancer. Previous studies demonstrated that inhibition of the MAPK pathway with a MEK inhibitor is synergistic with immune checkpoint inhibitors. PATIENTS AND METHODS We conducted a phase I/II trial of pembrolizumab and binimetinib in patients with metastatic triple-negative breast cancer with ≤3 prior lines of therapy. There were two dose levels (DL) with binimetinib at 45 mg at DL 0 and 30 mg at DL -1. RESULTS The recommended phase II dose was the standard dose of pembrolizumab with binimetinib 30 mg twice daily. The objective response rate (ORR) was 30.4%, with a numerically higher ORR in patients without liver metastasis at 45.5%. Among patients who achieved objective responses, 80% had a duration of response >12 months and ongoing even after stopping treatment (5.4-69.0 months). Patients with PD-L1-positive tumors (modified proportion score ≥10) were more likely to respond with an ORR of 66.7%. However, clinical benefit was observed in 25% of patients with PD-L1-negative tumors. Consistent with preclinical studies, four of six patients with clinical benefit had either increased PD-L1 or decreased p-ERK expressions in serial circulating cancer-associated macrophage-like cells after starting binimetinib. CONCLUSIONS Pembrolizumab and binimetinib at 30 mg are safe with manageable toxicities. Promising activity was observed in patients without liver metastases. Future larger clinical trials are warranted to further evaluate the efficacy of this chemotherapy-free combination.
Collapse
Affiliation(s)
- Saranya Chumsri
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Joseph J. Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Emily Liu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Kathleen S. Tenner
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | | | - Morgan T. Weidner
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Amanda N. Arnold
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Dana L. Haley
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Pooja Advani
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Kostandinos Sideras
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Alvaro Moreno-Aspitia
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | | | - Edith A. Perez
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Keith L. Knutson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
- Department of Immunology, Clinical and Research Laboratories, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
2
|
Feng H, Zhao LY, Xu Z, Xie QF, Deng HJ, Yu J, Liu H. Homologous recombination deficiency and immunotherapy response in microsatellite-stable colorectal cancer: Evidence from a cohort study in China. World J Gastrointest Oncol 2025; 17:102767. [DOI: 10.4251/wjgo.v17.i5.102767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/14/2025] [Accepted: 03/07/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Patients with colorectal cancer (CRC) exhibiting microsatellite instability (MSI)-high generally demonstrate a favorable response to immunotherapy. In contrast, the efficacy of immunotherapy in microsatellite-stable (MSS) CRC patients is considerably restricted. This study sought to evaluate the effectiveness of immunotherapy in MSS patients characterized by homologous recombination deficiency (HRD) as opposed to those with homologous recombination proficiency (HRP).
AIM To investigate and compare the clinicopathological characteristics, treatment modalities, and outcomes between the HRD and HRP groups in CRC.
METHODS Next-generation sequencing was performed on 268 CRC patients to identify tumor-associated genetic alterations and assess their HRD scores and MSI status. Patients with HRD-related gene alterations or an HRD score ≥ 30 were classified into the HRD group, while the remaining patients were assigned to the HRP group. Clinical data, including staging and treatment regimens, were collected for analysis. Cox regression and Kaplan-Meier survival curves were employed to evaluate whether the HRD group demonstrated improved survival outcomes following immunotherapy treatment.
RESULTS Among the 268 patients, 64 were classified into the HRD group, which had a higher proportion of early-stage CRC diagnoses compared to the HRP group. Kaplan-Meier survival curves indicated significantly better survival rates in the HRD group compared to the HRP group across all cohorts, as well as among MSS patients treated with immunotherapy (P < 0.05).
CONCLUSION This study demonstrates that CRC patients with HRD have a more favorable prognosis and suggests that HRD status could serve as a predictive marker for immunotherapy response in MSS patients.
Collapse
Affiliation(s)
- Hao Feng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Li-Ying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Zhou Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Qing-Feng Xie
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hai-Jun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
3
|
Li L, Pu H, Zhang X, Guo X, Li G, Zhang M. Resistance to PD-1/PD-L1 immune checkpoint blockade in advanced non-small cell lung cancer. Crit Rev Oncol Hematol 2025; 209:104683. [PMID: 40024354 DOI: 10.1016/j.critrevonc.2025.104683] [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: 06/06/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Lung cancer is one of the most common malignant tumors, of which non-small cell lung cancer (NSCLC) accounts for about 85 %. Although immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 inhibitors, have significantly improved the prognosis of patients with NSCLC. There are still many patients do not benefit from ICIs. Primary resistance remains a major challenge in advanced NSCLC. The cancer-immunity cycle describes the process from antigen release to T cell recognition and killing of the tumor, which provides a framework for understanding anti-tumor immunity. The classical cycle consists of seven steps, and alterations at each stage can result in resistance. This review examines the current status of PD-1/PD-L1 blockade in the treatment of advanced NSCLC and explores potential mechanisms of resistance. We summarize the latest clinical trials of PD-1/PD-L1 inhibitors combined with other therapies and explore potential targets for overcoming primary resistance to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Haihong Pu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaoxin Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaotian Guo
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Guangrui Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
4
|
Block MS, Nelson GD, Chen J, Johnson S, Yang L, Flotte TJ, Grewal EP, McWilliams RR, Kottschade LA, Domingo-Musibay E, Markovic SN, Dimou A, Montane HN, Piltin MA, Price DL, Khariwala SS, Hui JYC, Erskine CL, Strand CA, Zahrieh D, Dong H, Hieken TJ. Neoadjuvant cobimetinib and atezolizumab with or without vemurafenib for stage III melanoma: outcomes and the impact of the microbiome from the NeoACTIVATE trial. J Immunother Cancer 2025; 13:e011706. [PMID: 40234093 PMCID: PMC12001372 DOI: 10.1136/jitc-2025-011706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Neoadjuvant treatment has become standard for patients with high-risk operable stage III melanoma, but the optimal regimen is unknown. Targeted therapy approaches yield high pathological response rates, while immunotherapy regimens show favorable recurrence-free survival (RFS). NeoACTIVATE was designed to address whether a neoadjuvant combination of both targeted therapy and immunotherapy might leverage the benefits of each. METHODS We tested neoadjuvant treatment with 12 weeks of vemurafenib, cobimetinib, and atezolizumab for patients with BRAF-mutated (BRAFm) melanoma (cohort A) and cobimetinib and atezolizumab for patients with BRAF-wild-type (BRAFwt) melanoma (cohort B), regimens which we have shown generate a substantial major pathological response. After therapeutic lymph node dissection, patients received 24 weeks of adjuvant atezolizumab. Here, we report survival outcomes and their association with biomarkers assayed among the gut microbiome and peripheral blood immune subsets. RESULTS With 49 months median follow-up, the median RFS was not reached for cohort A and was 40.8 months for cohort B. At 24 months after operation, 2 of 14 cohort A patients and 4 of 13 cohort B patients had experienced distant relapse. Key findings from correlative analyses included diversity, taxonomic and functional metagenomic gut microbiome signals associated with distant metastasis-free survival at 2 years. Notably, we observed a strong correlation between low microbial arginine biosynthesis (required for T-cell activation and effector function) and early distant recurrence (p=0.0005), which correlated with taxonomic differential abundance findings. Peripheral blood immune monitoring revealed increased double-positive (CD4+CD8+) T cells in patients with early recurrence. CONCLUSIONS Neoadjuvant treatment with cobimetinib and atezolizumab±vemurafenib was associated with a low rate of distant metastasis in patients with high-risk stage III melanoma. Freedom from early distant metastasis was highly associated with taxonomic differences in gut microbiome structure and with functional pathway alterations known to modulate T cell immunity. Identification of predictive biomarkers will permit optimization of neoadjuvant therapy regimens for individual patients. TRIAL REGISTRATION NUMBER NCT03554083.
Collapse
Affiliation(s)
- Matthew S Block
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Garth D Nelson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen Johnson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Lu Yang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas James Flotte
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric P Grewal
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Evidio Domingo-Musibay
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | - Mara A Piltin
- Division of Breast and Melanoma Surgical Oncology, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel L Price
- Department of Otolaryngology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samir S Khariwala
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jane Yuet Ching Hui
- Division of Surgical Oncology, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | | | - Carrie A Strand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - David Zahrieh
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tina J Hieken
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Wang X, Liao Y, Liu D, Zheng J, Shi M. Presetting CAR-T cells during ex vivo biomanufacturing. Mol Ther 2025; 33:1380-1406. [PMID: 39988874 PMCID: PMC11997485 DOI: 10.1016/j.ymthe.2025.02.031] [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: 07/08/2024] [Revised: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the treatment of hematologic malignancies. However, it continues to encounter significant obstacles, including treatment relapse and limited efficacy in solid tumors. While effector T cells exhibit robust cytotoxicity, central memory T cells and stem cell-like T cells are essential for in vivo expansion, long-term survival, and persistence. Strategies such as genetic engineering to enhance CAR-T cell efficacy and durability are often accompanied by increased safety risks, which not only raise regulatory approval thresholds but also escalate CAR-T production costs. In contrast, optimizing ex vivo manufacturing conditions represents a more straightforward and practical approach, offering the potential for rapid application to commercially approved CAR-T products and enhancement of their clinical outcomes. This review examines several factors that have been shown to improve T cell memory phenotype and in vivo cytotoxic activity, including cytokines, electrolytes, signaling pathway inhibitors, metabolic modulators, and epigenetic agents. The insights provided will guide the optimization of CAR-T cell industrial production. Furthermore, considerations for selecting appropriate conditions are discussed, balancing effectiveness, cost-efficiency, safety, and regulatory compliance while addressing current challenges in the field.
Collapse
Affiliation(s)
- Xu Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ying Liao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Dan Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
6
|
Tak E, An HI, Lee AS, Han K, Choi J, Kim HD, Hong YS, Kim SY, Choi EK, Kim JE, Kim TW. Antitumor effects of immunotherapy combined with BRAF and MEK inhibitors in BRAF V600E metastatic colorectal cancer. Cancer Immunol Immunother 2025; 74:154. [PMID: 40105971 PMCID: PMC11923341 DOI: 10.1007/s00262-025-04005-3] [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: 07/11/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
BRAF-mutated colorectal cancer correlates with poor prognosis and limited response to standard treatments. Combining immune checkpoint inhibitors with BRAF/MEK inhibitors shows promise against BRAF-mutant melanoma in both preclinical and clinical trials. Therefore, we hypothesized that the treatment would be effective against BRAF-mutant colorectal cancer. In this study, we assessed the efficacy of combining immune checkpoint inhibitors with BRAF and/or MEK inhibitors in BRAF-mutant colorectal cancers. We treated BRAF V600E colorectal cancer cells HT-29 and SNU-1235 with encorafenib (BRAF inhibitor) and binimetinib (MEK inhibitor) and assessed the degrees of MAPK inhibition, JAK/STAT inhibition, cell viability, apoptosis, and the expression of antigen presenting machinery. We also inoculated HT-29 cells into mice and treated them with an immune checkpoint inhibitor (durvalumab), encorafenib, and binimetinib for 4 weeks. We found that treatment with BRAF inhibitor, MEK inhibitor, or their combination led to significant tumor growth reduction, along with the MAPK and JAK/STAT pathway inhibition, antigen presenting machinery induction, and cytotoxic T cell activation. Our study demonstrates the potential effectiveness of combining immune checkpoint inhibitors with BRAF or MEK inhibitors for BRAF-mutated colorectal cancers.
Collapse
Affiliation(s)
- Eunyoung Tak
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hye-In An
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Amy Sinyoung Lee
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyuyoung Han
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiwan Choi
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyung-Don Kim
- Department of Oncology, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Yong Sang Hong
- Department of Oncology, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Sun Young Kim
- Department of Oncology, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Eun Kyung Choi
- Department of Radiation Oncology, Asan Preclinical Evaluation Center for Cancer TherapeutiX, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Eun Kim
- Department of Oncology, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| | - Tae Won Kim
- Department of Oncology, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer TherapeutiX, Asan Medical Center, Seoul, 05505, Republic of Korea
| |
Collapse
|
7
|
Wang Y, Wang Y, Gao H, Chen L, Zheng S, Chen Y, Shi H, Han A. Ezetimibe mediated RPS6KA2 inhibits colorectal cancer proliferation via PCSK9/MAPK signaling pathway. Cancer Treat Res Commun 2025; 43:100899. [PMID: 40112524 DOI: 10.1016/j.ctarc.2025.100899] [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: 02/01/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
To investigate the effect and molecular mechanism of ezetimibe on colorectal cancer (CRC), our study found that ezetimibe significantly inhibited the proliferation and progression of CRC. Further study showed that RPS6KA2 might be the target gene of ezetimibe treatment on CRC. RPS6KA2 expression was significantly lower in human CRC tissue samples and associated with T classification and vascular invasion of tumor cells. RPS6KA2 inhibited proliferation, migration, and invasion of CRC cells. The underlying mechanisms indicated that interaction between RPS6KA2 and PCSK9 was observed within the cytoplasmic compartment of CRC cells. RPS6KA2 suppressed PCSK9 and MAPK signaling pathway in CRC cells. BI-D1780 which is an inhibitor of RPS6KA2 increased PCSK9 and MAPK signaling pathway related proteins expression in SW620 cells. However, an inhibitor or stimulator of MAPK did not affect RPS6KA2 and PCSK9 expression, respectively. In vivo, CRC cells with RPS6KA2 or PCSK9 overexpression could inhibit or promote tumor growth and metastasis, respectively. PCSK9 promoted proliferation, migration, and invasion of CRC cells. PCSK9 expression was higher in human CRC samples and associated with N classification and TNM stage of CRC. In conclusion, our study firstly suggests that ezetimibe suppresses CRC progression by upregulating RPS6KA2 while downregulating PCSK9/MAPK signaling pathway.
Collapse
Affiliation(s)
- Yu Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Yuting Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Huabin Gao
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Lin Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Shuai Zheng
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Yongyu Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Huijuan Shi
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China.
| | - Anjia Han
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China.
| |
Collapse
|
8
|
Brady AE, Revu S, Wu D, Fisk H, Kone K, Lydecker A, Purser EJ, Smith N, Hilt ZT, Woodyear S, Caddy S, Gingras S, Rudd B, McGeachy MM. Humanizing a CD28 signaling domain affects CD8 activation, exhaustion and stem-like precursors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642460. [PMID: 40161835 PMCID: PMC11952375 DOI: 10.1101/2025.03.10.642460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
CD28 ligation provides critical signals that modulate activated T cell fate. In a human to mouse reverse-engineering approach, a single amino acid substitution adjacent to the C-terminal proline-rich domain created CD28A210P mice with enhanced signaling. CD28A210P mice experienced pro-inflammatory responses to CD28 superagonist antibody, analogous to severe cytokine storm induced in a human clinical trial, with a striking increase of activated CD8 T cells. In acute and chronic viral infections, early activation and expansion of CD28A210P CD8 effector T cells increased, with accelerated exhaustion in chronic infection. Mechanistically, CD28A210P enhanced JunB, IL-2, and inhibitory receptors driven by MEK1/2. Generation of CD28A210P stem-like progenitor (Tpex) cells was enhanced in acute and chronic infections, and further expanded by PD-L1 blockade in chronically-infected mice. Thus, 'humanized' PYAP mice reveal key roles for CD28 signaling strength in CD8 activation, accelerating exhaustion during antigen persistence, while promoting and sustaining Tpex during acute and chronic viral infection.
Collapse
|
9
|
Fomin V, So WV, Barbieri RA, Hiller-Bittrolff K, Koletou E, Tu T, Gomes B, Cai J, Charo J. Machine learning identifies clinical tumor mutation landscape pathways of resistance to checkpoint inhibitor therapy in NSCLC. J Immunother Cancer 2025; 13:e009092. [PMID: 40032600 PMCID: PMC11877243 DOI: 10.1136/jitc-2024-009092] [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: 03/11/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (CPIs) have revolutionized cancer therapy for several tumor indications. However, a substantial fraction of patients treated with CPIs derive no benefit or have short-lived responses to CPI therapy. Identifying patients who are most likely to benefit from CPIs and deciphering resistance mechanisms is therefore essential for developing adjunct treatments that can abrogate tumor resistance. PATIENTS AND METHODS In this study, we used a machine learning approach that used the US-based nationwide de-identified Flatiron Health and Foundation Medicine non-small cell lung carcinoma (NSCLC) clinico-genomic database to identify genomic markers that predict clinical responses to CPI therapy. In total, we analyzed data from 4,433 patients with NSCLC. RESULTS Analysis of pretreatment genomic data from 1,511 patients with NSCLC identified. Of the 36 genomic signatures identified, 33 exhibited strong predictive capacity for CPI response (n=1150) compared with chemotherapy response (n=361), while three signatures were prognostic. These 36 genetic signatures had in common a core set of four genes (BRAF, BRIP1, FGF10, and FLT1). Interestingly, we observed that some (n=19) of the genes in the signatures (eg, TP53, EZH2, KEAP1 and FGFR2) had alternative mutations with contrasting clinical outcomes to CPI therapy. Finally, the genetic signatures revealed multiple biological pathways involved in CPI response, including MAPK, PDGF, IL-6 and EGFR signaling. CONCLUSIONS In summary, we found several genomic markers and pathways that provide insight into biological mechanisms affecting response to CPI therapy. The analyses identified novel targets and biomarkers that have the potential to provide candidates for combination therapies or patient enrichment strategies, which could increase response rates to CPI therapy in patients with NSCLC.
Collapse
Affiliation(s)
- Vitalay Fomin
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
- Numenos, New York, NY, USA
| | - WeiQing Venus So
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
| | | | | | - Elina Koletou
- Roche Pharmaceutical Research and Early Development, Data and Analytics, Roche Innovation Center Basel, Basel, Switzerland
| | - Tiffany Tu
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
| | - Bruno Gomes
- Roche Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - James Cai
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
| | - Jehad Charo
- Roche Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| |
Collapse
|
10
|
Viel S, Vivier E, Walzer T, Marçais A. Targeting metabolic dysfunction of CD8 T cells and natural killer cells in cancer. Nat Rev Drug Discov 2025; 24:190-208. [PMID: 39668206 DOI: 10.1038/s41573-024-01098-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 12/14/2024]
Abstract
The importance of metabolic pathways in regulating immune responses is now well established, and a mapping of the bioenergetic metabolism of different immune cell types is under way. CD8 T cells and natural killer (NK) cells contribute to cancer immunosurveillance through their cytotoxic functions and secretion of cytokines and chemokines, complementing each other in target recognition mechanisms. Several immunotherapies leverage these cell types by either stimulating their activity or redirecting their specificity against tumour cells. However, the anticancer activity of CD8 T cells and NK cells is rapidly diminished in the tumour microenvironment, closely linked to a decline in their metabolic capacities. Various strategies have been developed to restore cancer immunosurveillance, including targeting bioenergetic metabolism or genetic engineering. This Review provides an overview of metabolic dysfunction in CD8 T cells and NK cells within the tumour microenvironment, highlighting current therapies aiming to overcome these issues.
Collapse
Affiliation(s)
- Sébastien Viel
- Plateforme de Biothérapie et de Production de Médicaments de Thérapie Innovante, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
- APHM, Hôpital de la Timone, Marseille, France
- Paris Saclay Cancer Cluster, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Inserm, Prédicteurs moléculaires et nouvelles cibles en oncologie, Villejuif, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France.
| |
Collapse
|
11
|
Ramos-Ramírez M, Caballe-Pérez E, Lucio-Lozada J, Romero-Nuñez E, Castillo-Ruiz C, Dorantes-Sánchez L, Flores-Estrada D, Recondo G, Barrios-Bernal P, Cabrera-Miranda L, Bravo-Dominguez H, Hernández-Pedro N, Arrieta O. Immunomodulatory role of oncogenic alterations in non-small cell lung cancer: a review of implications for immunotherapy. Cancer Metastasis Rev 2025; 44:30. [PMID: 39915358 DOI: 10.1007/s10555-025-10245-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/16/2025] [Indexed: 03/28/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved clinical outcomes in patients with non-small cell lung cancer (NSCLC) lacking targetable oncogenic alterations. However, their efficacy in individuals with such genomic alterations remains heterogeneous and poorly understood. In detail, certain oncogenic alterations in TP53, EGFR (uncommon mutations), KRAS (G12C), BRAF (non-V600E), MET (amplifications), FGFR1 and FGFR4, actively modify MAPK, PI3K, and STING signaling, thus remodeling tumoral immune phenotype and are associated with high TMB counts, enriched T lymphocyte tumor infiltration, and high expression of antigen-presenting molecules, supporting their consideration as part of the eligibility criteria for ICIs treatment. Nonetheless, other oncogenic alterations are associated with an immunosuppressive TME, low TMB counts, and downregulation of targetable immune checkpoints, in which novel therapeutic approaches are currently being tested to overcome their intrinsic resistance. In this context, this review discusses the fundamental mechanisms by which frequent driver alterations affect ICIs efficacy in patients with NSCLC, and outlines their prognostic relevance in the era of immunotherapy.
Collapse
Affiliation(s)
- Maritza Ramos-Ramírez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Enrique Caballe-Pérez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - José Lucio-Lozada
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Eunice Romero-Nuñez
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Cesar Castillo-Ruiz
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Lorena Dorantes-Sánchez
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Diana Flores-Estrada
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Gonzalo Recondo
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pedro Barrios-Bernal
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Luis Cabrera-Miranda
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Heyman Bravo-Dominguez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Norma Hernández-Pedro
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico.
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico.
| | - Oscar Arrieta
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico.
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico.
| |
Collapse
|
12
|
Kim Y, Song J, Kim N, Sim T. Recent progress in emerging molecular targeted therapies for intrahepatic cholangiocarcinoma. RSC Med Chem 2025:d4md00881b. [PMID: 39925737 PMCID: PMC11800140 DOI: 10.1039/d4md00881b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/11/2025] [Indexed: 02/11/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a diverse group of epithelial malignant tumors arising from the biliary tract, characterized by high molecular heterogeneity. It is classified into intrahepatic (iCCA) and extrahepatic CCA (eCCA) based on the location of the primary tumor. CCA accounts for approximately 15% of all primary liver cancers, with iCCA comprising 10-20% of all CCAs. iCCA is especially known for its characteristic aggressiveness and refractoriness, leading to poor prognosis. Despite the increasing global incidence and mortality rates, surgery remains the only available standard treatment approach for a subset (25%) of patients with early-stage, resectable iCCA. The paucity of effective systemic medical therapies restricts therapeutic options for patients with advanced or metastatic iCCA. In the past decade, advances in the understanding of the molecular complexity of these tumors have provided fruitful insights for the identification of promising new druggable targets and the development of feasible therapeutic strategies that may improve treatment outcomes for patients with iCCA. In this review, we aim to highlight critical up-to-date studies and medicinal chemistry aspects, focusing on novel targeted approaches utilizing promising candidates for molecular targeted therapy in iCCA. These candidates include aberrations in isocitrate dehydrogenase (IDH) 1/2, fibroblast growth factor receptor (FGFR), B-Raf proto-oncogene (BRAF), neurotrophic tyrosine receptor kinase (NTRK), human epidermal growth factor receptor 2 (HER2), and programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1). Furthermore, this review provides an overview of potential inhibitors aimed at overcoming acquired drug resistance in these actionable targets for iCCA.
Collapse
Affiliation(s)
- Younghoon Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
- Clinical Candidate Discovery & Development Institute, Yonsei University College of Medicine Seoul Korea
| | - Jaewon Song
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
| | - Namkyoung Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
- Clinical Candidate Discovery & Development Institute, Yonsei University College of Medicine Seoul Korea
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
| |
Collapse
|
13
|
Gorría T, Sierra-Boada M, Rojas M, Figueras C, Marin S, Madurga S, Cascante M, Maurel J. Metabolic Singularities in Microsatellite-Stable Colorectal Cancer: Identifying Key Players in Immunosuppression to Improve the Immunotherapy Response. Cancers (Basel) 2025; 17:498. [PMID: 39941865 PMCID: PMC11815897 DOI: 10.3390/cancers17030498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Although immune checkpoint inhibitor (ICI) therapy is currently the standard of care in microsatellite-unstable (MSI) metastatic colorectal cancer (CRC), ICI therapy, alone or in combination with other therapies, is not a treatment approach in microsatellite-stable (MSS) CRC, which is present in 95% of patients. In this review, we focus on metabolic singularities-at the transcriptomic (either bulk or single cell), proteomic, and post-translational modification levels-that induce immunosuppression in cancer and specifically in MSS CRC. First, we evaluate the current efficacy of ICIs in limited and metastatic disease in MSS CRC. Second, we discuss the latest findings on the potential biomarkers for evaluating ICI efficacy in MSS CRC using strict REMARK criteria. Third, we review the current evidence on metabolic patterns in CRC tumors and immune cell metabolism to advance our understanding of metabolic crosstalk and to pave the way for the development of combination strategies to enhance ICI efficacy.
Collapse
Affiliation(s)
- Teresa Gorría
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
- Translational Genomics and Targeted Therapies in Solid Tumors, Agustí Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
| | - Marina Sierra-Boada
- Medical Oncology Department, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain;
| | - Mariam Rojas
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
| | - Carolina Figueras
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08036 Barcelona, Spain;
- Institute of Biomedicine of University of Barcelona (IBUB), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Sergio Madurga
- Department of Material Science and Physical Chemistry, Research Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, 08028 Barcelona, Spain;
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08036 Barcelona, Spain;
- Institute of Biomedicine of University of Barcelona (IBUB), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Joan Maurel
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
- Translational Genomics and Targeted Therapies in Solid Tumors, Agustí Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
14
|
Foffano L, Bertoli E, Bortolot M, Torresan S, De Carlo E, Stanzione B, Del Conte A, Puglisi F, Spina M, Bearz A. Immunotherapy in Oncogene-Addicted NSCLC: Evidence and Therapeutic Approaches. Int J Mol Sci 2025; 26:583. [PMID: 39859299 PMCID: PMC11765476 DOI: 10.3390/ijms26020583] [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: 12/20/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. The discovery of specific driver mutations has revolutionized the treatment landscape of oncogene-addicted NSCLC through targeted therapies, significantly improving patient outcomes. However, immune checkpoint inhibitors (ICIs) have demonstrated limited effectiveness in this context. Emerging evidence, though, reveals significant heterogeneity among different driver mutation subgroups, suggesting that certain patient subsets may benefit from ICIs, particularly when combined with other therapeutic modalities. In this review, we comprehensively examine the current evidence on the efficacy of immunotherapy in oncogene-addicted NSCLC. By analyzing recent clinical trials and preclinical studies, along with an overview of mechanisms that may reduce immunotherapy efficacy, we explored potential strategies to address these challenges, to provide insights that could optimize immunotherapy approaches and integrate them effectively into the treatment algorithm for oncogene-addicted NSCLC.
Collapse
Affiliation(s)
- Lorenzo Foffano
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Martina Bortolot
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Sara Torresan
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Brigida Stanzione
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Alessandro Del Conte
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Fabio Puglisi
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Michele Spina
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Alessandra Bearz
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| |
Collapse
|
15
|
Gu Y, Yang R, Zhang Y, Guo M, Takehiro K, Zhan M, Yang L, Wang H. Molecular mechanisms and therapeutic strategies in overcoming chemotherapy resistance in cancer. MOLECULAR BIOMEDICINE 2025; 6:2. [PMID: 39757310 PMCID: PMC11700966 DOI: 10.1186/s43556-024-00239-2] [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: 05/31/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Cancer remains a leading cause of mortality globally and a major health burden, with chemotherapy often serving as the primary therapeutic option for patients with advanced-stage disease, partially compensating for the limitations of non-curative treatments. However, the emergence of chemotherapy resistance significantly limits its efficacy, posing a major clinical challenge. Moreover, heterogeneity of resistance mechanisms across cancer types complicates the development of universally effective diagnostic and therapeutic approaches. Understanding the molecular mechanisms of chemoresistance and identifying strategies to overcome it are current research focal points. This review provides a comprehensive analysis of the key molecular mechanisms underlying chemotherapy resistance, including drug efflux, enhanced DNA damage repair (DDR), apoptosis evasion, epigenetic modifications, altered intracellular drug metabolism, and the role of cancer stem cells (CSCs). We also examine specific causes of resistance in major cancer types and highlight various molecular targets involved in resistance. Finally, we discuss current strategies aiming at overcoming chemotherapy resistance, such as combination therapies, targeted treatments, and novel drug delivery systems, while proposing future directions for research in this evolving field. By addressing these molecular barriers, this review lays a foundation for the development of more effective cancer therapies aimed at mitigating chemotherapy resistance.
Collapse
Affiliation(s)
- Yixiang Gu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ruifeng Yang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yang Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Miaomiao Guo
- The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | | | - Ming Zhan
- The Core Laboratory in Medical Center of Clinical Research, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Department of Systems Biology, Beckman Research Institute, City of Hope, Monrovia, CA, 91016, USA
| | - Linhua Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Hui Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
16
|
Ryan C, Moore A, Davis M, Yazel C, Momtahen S, Ricci AM, Talbot EA, Mann J, Pace N. A rare cutaneous infection with Mycobacterium chelonae in a pediatric patient treated with trametinib for KRAS-mutated diffuse glioma. Pediatr Dermatol 2025; 42:121-124. [PMID: 39148304 DOI: 10.1111/pde.15718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/20/2024] [Indexed: 08/17/2024]
Abstract
Mitogen-activated extracellular signal-regulated kinase inhibitors (MEKi) represent a promising new therapy for pediatric patients with low-grade gliomas, which frequently have abnormal signaling within the mitogen-activated protein kinase (MAP kinase) pathway. However, understanding of long-term efficacy and toxicity is limited in pediatric glioma patients. This article describes a rare presentation of a widespread cutaneous infection with Mycobacterium chelonae in a pediatric patient with a low-grade glioma treated with trametinib.
Collapse
Affiliation(s)
- Chenin Ryan
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Angel Moore
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Matthew Davis
- Department of Dermatology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Caitlin Yazel
- Department of Dermatology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Shabnam Momtahen
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Angela M Ricci
- Section of Pediatric Hematology/Oncology, Department of Pediatrics, Children's Hospital at Dartmouth-Hitchcock, Lebanon, New Hampshire, USA
| | - Elizabeth A Talbot
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Infectious Disease and International Health, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Julianne Mann
- Section of Dermatology, Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Nicole Pace
- Section of Dermatology, Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
17
|
Yao Y, Wu M, Wang Y, Liao Z, Yang Y, Liu Y, Shi J, Wu W, Wei X, Xu J, Guo Y, Dong X, Che J, Wang J, Gu Z. An Oral PROTAC Targeting HPK1 Degradation Potentiates Anti-Solid Tumor Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411454. [PMID: 39568237 DOI: 10.1002/adma.202411454] [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: 08/04/2024] [Revised: 09/25/2024] [Indexed: 11/22/2024]
Abstract
Hematopoietic progenitor pinase1 (HPK1) knockout has been identified as an efficient route to enhance anti-tumor immune response. Here, this work develops an oral proteolysis targeting chimera (PROTAC) targeting HPK1 to efficiently and selectively degrade HPK1 to augment immunotherapeutic outcomes. In a postoperative tumor model of human cervical cancer in NSG mice, the orally-administrated PROTAC can reach tumors, down-regulate HPK1 levels in locally-administrated CAR-T cells, and promote their efficiency in inhibiting solid tumor recurrence, achieving 50% partial response (PR) and 50% complete response (CR). In addition, oral administration of PROTAC can amplify the suppression capability of the anti-PD-L1 antibody on the growth of CT26 solid tumors in BALB/c mice by promoting the infiltration of CD45-positive immune cells from 0.7% to 1.5% and CD3-positive T cells from 0.2% to 0.5% within the tumors.
Collapse
Affiliation(s)
- Yuejun Yao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Mingfei Wu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanfang Wang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Ziyan Liao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Yinxian Yang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Yun Liu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jiaqi Shi
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xinwei Wei
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jianchang Xu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Yugang Guo
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaowu Dong
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jinxin Che
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jinqiang Wang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhen Gu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310000, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310029, China, Liangzhu Laboratory, Hangzhou, 311121, China
| |
Collapse
|
18
|
Gong Y, Kang J, Wang M, Hayati F, Syed Abdul Rahim SS, Poh Wah Goh L. The trends and hotspots of immunotherapy for metastatic colorectal cancer from 2013 to 2022: A bibliometric and visual analysis. Hum Vaccin Immunother 2024; 20:2312599. [PMID: 38356280 PMCID: PMC10877983 DOI: 10.1080/21645515.2024.2312599] [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: 11/12/2023] [Accepted: 01/27/2024] [Indexed: 02/16/2024] Open
Abstract
An increasing body of research indicates that immunotherapy has demonstrated substantial effectiveness in the realm of metastatic colorectal cancer(mCRC), especially among patients with deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) (dMMR/MSI-H mCRC). This study constitutes the inaugural bibliometric and visual analysis of immunotherapy related to mCRC during the last decade. Between 2013 and the conclusion of 2022, we screened 306 articles from Web of Science and subjected them to analysis using CiteSpace and VOSviewer. The United States stood out as the primary contributor in this area, representing 33.33% of the publications, with China following closely at 24.51%. The most prolific institution has the lowest average citation rate. Sorbonne University were the most highly cited institutions. Notably, Frontiers In Oncology published the largest quantity of articles. Andre, Thierry, and Overman, Michael J. were prominent authors known for their prolific output and the high citation rates of their work. The focus areas in this field encompass "tumor microenvironment," "liver metastasis," "tumor-associated macrophages," "combination therapy" and "gut microbiota." Some keywords offer promise as potential biomarkers for evaluating the effectiveness of immunotherapeutic interventions.
Collapse
Affiliation(s)
- Yifan Gong
- Faculty of Medicine and Health Science, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| | - Jianping Kang
- Orthopedics Ward 2, Yunnan Cancer Hospital, Kunming, China
| | - Mingting Wang
- Oncology Department, Affiliated Hospital of Panhihua University, Panzhihua, China
| | - Firdaus Hayati
- Faculty of Medicine and Health Science, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| | | | - Lucky Poh Wah Goh
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| |
Collapse
|
19
|
Qu F, Wu S, Yu W. Progress of Immune Checkpoint Inhibitors Therapy for pMMR/MSS Metastatic Colorectal Cancer. Onco Targets Ther 2024; 17:1223-1253. [PMID: 39735789 PMCID: PMC11681808 DOI: 10.2147/ott.s500281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 12/31/2024] Open
Abstract
Immunotherapy is one of the research hotspots in colorectal cancer field in recent years. The colorectal cancer patients with mismatch repair-deficient (dMMR) or high microsatellite instability (MSI-H) are the primary beneficiaries of immunotherapy. However, the vast majority of colorectal cancers are mismatch repair proficient (pMMR) or microsatellite stability (MSS), and their immune microenvironment is characterized by "cold tumors" that are generally insensitive to single immunotherapy based on immune checkpoint inhibitors (ICIs). Studies have shown that some pMMR/MSS colorectal cancer patients regulate the immune microenvironment by combining other treatments, such as multi-target tyrosine kinase inhibitors, anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, chemotherapy, radiotherapy, anti-epithelial growth factor receptor (EGFR) monoclonal antibodies, and mitogen-activated protein kinase (MAPK) signaling pathway inhibitors and oncolytic viruses, etc. to transform "cold tumor" into "hot tumor", thereby improving the response to immunotherapy. In addition, screening for potential prognostic biomarkers can also enrich the population benefiting from immunotherapy for microsatellite stable colorectal cancer. Therefore, in pMMR or MSS metastatic colorectal cancer (mCRC), the optimization of immunotherapy regimens and the search for effective efficacy prediction biomarkers are currently important research directions. In this paper, we review the progress of efficacy of immunotherapy (mainly ICIs) in pMMR /MSS mCRC, challenges and potential markers, in order to provide research ideas for the development of immunotherapy for mCRC.
Collapse
Affiliation(s)
- Fanjie Qu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - Shuang Wu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - WeiWei Yu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| |
Collapse
|
20
|
Cui C, Zhang H, Yang C, Yin M, Teng X, Yang M, Kong D, Zhang J, Peng W, Chu Z, Wang J, Sun Y, Kang L, Lyu B, Gao Q, Wu M, Wang Y, Li Y. Inhibition of JNK Signaling Overcomes Cancer-Associated Fibroblast-Mediated Immunosuppression and Enhances the Efficacy of Immunotherapy in Bladder Cancer. Cancer Res 2024; 84:4199-4213. [PMID: 39292817 DOI: 10.1158/0008-5472.can-24-0940] [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: 03/29/2024] [Revised: 07/15/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024]
Abstract
Currently, only 20% to 40% of patients with cancer benefit from immune checkpoint inhibitors. Understanding the mechanisms underlying the immunosuppressive tumor microenvironment (TME) and characterizing dynamic changes in the immunologic landscape during treatment are critical for improving responsiveness to immunotherapy. In this study, we identified JNK signaling in cancer-associated fibroblasts (CAF) as a regulator of the immunosuppressive TME. Single-cell RNA sequencing of bladder cancer samples treated with a JNK inhibitor revealed enhanced cytotoxicity and effector functions of CD8+ T cells. In untreated tumors, CAFs interacted frequently with CD8+ T cells and mediated their exhaustion. JNK inhibition abrogated the immunosuppression function of CAFs by downregulating the expression of thymic stromal lymphopoietin (TSLP), thereby restoring CD8+ T-cell cytotoxicity. In addition, blockade of CAF-derived TSLP in combination with anti-PD-1 treatment promoted tumor elimination by CD8+ T cells in vivo. Collectively, these results indicate that JNK signaling plays an important immunosuppressive role in the TME by promoting expression of TSLP in CAFs and suggest that inhibiting JNK signaling could be a promising immunotherapeutic strategy for cancer treatment. Significance: JNK signaling promotes the secretion of TSLP by bladder cancer-associated fibroblasts to impede CD8+ T-cell activity, which can be circumvented by combination treatment targeting JNK signaling and PD-1.
Collapse
Affiliation(s)
- Chengying Cui
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Haojie Zhang
- Department of Urology, Huadong Hospital, Fudan University, Shanghai, China
| | - Congcong Yang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Mingwei Yin
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinkun Teng
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Miaomiao Yang
- The First Affiliated Hospital of Anhui Medical University, Pathology Center, Hefei, China
- Anhui Public Health Clinical Center, Pathology Center, Hefei, China
| | - Dejie Kong
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Jinzhi Zhang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Weidong Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Zhimin Chu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Jingjing Wang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Yating Sun
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Liping Kang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Bin Lyu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Qian Gao
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Mingqing Wu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Yongqiang Wang
- Department of Urology, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| | - Yang Li
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Bortolot M, Torresan S, De Carlo E, Bertoli E, Stanzione B, Del Conte A, Spina M, Bearz A. Navigating Therapeutic Challenges in BRAF-Mutated NSCLC: Non-V600 Mutations, Immunotherapy, and Overcoming Resistance. Int J Mol Sci 2024; 25:12972. [PMID: 39684685 DOI: 10.3390/ijms252312972] [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: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Although rare in non-small cell lung cancer (NSCLC), BRAF mutations present considerable therapeutic challenges. While the use of BRAF and MEK inhibitor combinations has significantly improved survival outcomes in patients with BRAF V600E mutations, no targeted therapies are currently available for class II and III mutations, leaving the optimal treatment strategy and prognosis for these patients uncertain. Additionally, despite immunotherapy typically showing limited benefit in patients with other activating genomic alterations, it appears to deliver comparable efficacy in BRAF-mutated NSCLC, emerging as a potentially viable treatment option, particularly in patients with a history of smoking. However, resistance to BRAF pathway inhibitors is inevitable, leading to disease progression, and a well-defined strategy to overcome these resistance mechanisms is lacking. This review aims to explore the critical challenges in the management of BRAF-mutated NSCLC, providing a comprehensive summary of the current evidence and highlighting ongoing clinical trials that aim to address these critical gaps.
Collapse
Affiliation(s)
- Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Sara Torresan
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
22
|
Liu M, Liu Q, Hu K, Dong Y, Sun X, Zou Z, Ji D, Liu T, Yu Y. Colorectal cancer with BRAF V600E mutation: Trends in immune checkpoint inhibitor treatment. Crit Rev Oncol Hematol 2024; 204:104497. [PMID: 39245296 DOI: 10.1016/j.critrevonc.2024.104497] [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: 04/02/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024] Open
Abstract
Colorectal cancer (CRC) with BRAF V600E mutation presents a formidable scientific and clinical challenge due to its aggressive nature and poor response to standard therapeutic approaches. BRAF V600E mutation-induced conspicuous activation of the MAPK pathway contributes to the relentless tumor progression. Nevertheless, the efficacy of multi-targeted MAPK pathway inhibition remains suboptimal in clinical practice. Patients with high microsatellite instability (MSI-H) have shown favorable results with immune checkpoint inhibitors (ICIs). The combination of the MAPK pathway inhibition with ICIs has recently emerged as a promising regimen to improve clinical outcomes in the microsatellite stable (MSS) subgroup of BRAF V600E-mutant metastatic CRC patients. In this review, we elucidate the unique tumor biology of BRAF V600E-mutant CRC, with a particular focus on the immune features underlying the rationale for ICI treatments in the MSI-H and MSS subpopulations, then highlight the trends in clinical trials of the ICI therapy for BRAF V600E-mutant metastatic CRC.
Collapse
Affiliation(s)
- Mengling Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Keshu Hu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Dong
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xun Sun
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhiguo Zou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dingkun Ji
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
23
|
Maggi E, Munari E, Landolina N, Mariotti FR, Azzarone B, Moretta L. T cell landscape in the microenvironment of human solid tumors. Immunol Lett 2024; 270:106942. [PMID: 39486594 DOI: 10.1016/j.imlet.2024.106942] [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: 10/03/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
T cells are the main effectors involved in anti-tumor immunity, mediating most of the adaptive response towards cancer. After priming in lymph nodes, tumor antigens-specific naïve T lymphocytes proliferate and differentiate into effector CD4+ and CD8+ T cells that migrate from periphery into tumor sites aiming to eliminate cancer cells. Then while most effector T cells die, a small fraction persists and recirculates as long-lived memory T cells which generate enhanced immune responses when re-encountering the same antigen. A number of T (and non-T) cell subsets, stably resides in non-lymphoid peripheral tissues and may provide rapid immune response independently of T cells recruited from blood, against the reemergence of cancer cells. When tumor grows, however, tumor cells have evaded immune surveillance of effector cells (NK and CTL cells) which are exhausted, thus favoring the local expansion of T (and non-T) regulatory cells. In this review, the current knowledge of features of T cells present in the tumor microenvironment (TME) of solid adult and pediatric tumors, the mechanisms upregulating immune-checkpoint molecules and transcriptional and epigenetic landscapes leading to dysfunction and exhaustion of T effector cells are reviewed. The interaction of T cells with cancer- or TME non-neoplastic cells and their secreted molecules shape the T cell profile compromising the intrinsic plasticity of T cells and, therefore, favoring immune evasion. In this phase regulatory T cells contribute to maintain a high immunosuppressive TME thus facilitating tumor cell proliferation and metastatic spread. Despite the advancements of cancer immunotherapy, many tumors are unresponsive to immune checkpoint inhibitors, or therapeutical vaccines or CAR T cell-based adoptive therapy: some novel strategies to improve these T cell-based treatments are lastly proposed.
Collapse
Affiliation(s)
- Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Enrico Munari
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona 37126, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | | | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy.
| |
Collapse
|
24
|
Xie Q, Liu X, Liu R, Pan J, Liang J. Cellular mechanisms of combining innate immunity activation with PD-1/PD-L1 blockade in treatment of colorectal cancer. Mol Cancer 2024; 23:252. [PMID: 39529058 PMCID: PMC11555832 DOI: 10.1186/s12943-024-02166-w] [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: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
PD-1/PD-L1 blockade therapies have displayed extraordinary clinical efficacy for melanoma, renal, bladder and lung cancer; however, only a minority of colorectal cancer (CRC) patients benefit from these treatments. The efficacy of PD-1/PD-L1 blockade in CRC is limited by the complexities of tumor microenvironment. PD-1/PD-L1 blockade immunotherapy is based on T cell-centered view of tumor immunity. However, the onset and maintenance of T cell responses and the development of long-lasting memory T cells depend on innate immune responses. Acknowledging the pivotal role of innate immunity in anti-tumor immune response, this review encapsulates the employment of combinational therapies those involve PD-1/PD-L1 blockade alongside the activation of innate immunity and explores the underlying cellular mechanisms, aiming to harnessing innate immune responses to induce long-lasting tumor control for CRC patients who received PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
| |
Collapse
|
25
|
Wang X, Tao X, Chen P, Jiang P, Li W, Chang H, Wei C, Lai X, Zhang H, Pan Y, Ding L, Liang Z, Cui J, Shao M, Teng X, Gu T, Wei J, Kong D, Si X, Han Y, Fu H, Lin Y, Yu J, Li X, Wang D, Hu Y, Qian P, Huang H. MEK inhibition prevents CAR-T cell exhaustion and differentiation via downregulation of c-Fos and JunB. Signal Transduct Target Ther 2024; 9:293. [PMID: 39438476 PMCID: PMC11496645 DOI: 10.1038/s41392-024-01986-y] [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: 11/27/2023] [Revised: 08/26/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Clinical evidence supports the notion that T cell exhaustion and terminal differentiation pose challenges to the persistence and effectiveness of chimeric antigen receptor-T (CAR-T) cells. MEK1/2 inhibitors (MEKIs), widely used in cancer treatment due to their ability to inhibit aberrant MAPK signaling, have shown potential synergistic effects when combined with immunotherapy. However, the impact and mechanisms of MEKIs on CAR-T cells remain uncertain and controversial. To address this, we conducted a comprehensive investigation to determine whether MEKIs enhance or impair the efficacy of CAR-T cells. Our findings revealed that MEKIs attenuated CAR-T cell exhaustion and terminal differentiation induced by tonic signaling and antigen stimulation, thereby improving CAR-T cell efficacy against hematological and solid tumors. Remarkably, these effects were independent of the specific scFvs and costimulatory domains utilized in CARs. Mechanistically, analysis of bulk and single-cell transcriptional profiles demonstrates that the effect of MEK inhibition was related to diminish anabolic metabolism and downregulation of c-Fos and JunB. Additionally, the overexpression of c-Fos or JunB in CAR-T cells counteracted the effects of MEK inhibition. Furthermore, our Cut-and-Tag assay revealed that MEK inhibition downregulated the JunB-driven gene profiles associated with exhaustion, differentiation, anergy, glycolysis, and apoptosis. In summary, our research unveil the critical role of the MAPK-c-Fos-JunB axis in driving CAR-T cell exhaustion and terminal differentiation. These mechanistic insights significantly broaden the potential application of MEKIs to enhance the effectiveness of CAR-T therapy.
Collapse
Affiliation(s)
- Xiujian Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Xiao Tao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Pengjie Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Penglei Jiang
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
- Center of Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wenxiao Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Hefeng Chang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Cong Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Xinyi Lai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Hao Zhang
- Department of Hematology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Yihan Pan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Lijuan Ding
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Zuyu Liang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Jiazhen Cui
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Mi Shao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Xinyi Teng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Tianning Gu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Jieping Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Delin Kong
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Xiaohui Si
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Yingli Han
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Huarui Fu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Yu Lin
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Jian Yu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Xia Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Dongrui Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Pengxu Qian
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China.
- Center of Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China.
| |
Collapse
|
26
|
Rosenlehner T, Pennavaria S, Akçabozan B, Jahani S, O'Neill TJ, Krappmann D, Straub T, Kranich J, Obst R. Reciprocal regulation of mTORC1 signaling and ribosomal biosynthesis determines cell cycle progression in activated T cells. Sci Signal 2024; 17:eadi8753. [PMID: 39436996 DOI: 10.1126/scisignal.adi8753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 05/10/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024]
Abstract
Ribosomal biosynthesis in nucleoli is an energy-demanding process driven by all RNA polymerases and hundreds of auxiliary proteins. We investigated how this process is regulated in activated T lymphocytes by T cell receptor (TCR) signals and the multiprotein complexes mTORC1 and mTORC2, both of which contain the kinase mTOR. Deficiency in mTORC1 slowed the proliferation of T cells, with further delays in each consecutive division, an effect not seen with deficiency in mTORC2. mTORC1 signaling was stimulated by components of conventional TCR signaling, and, reciprocally, TCR sensitivity was decreased by mTORC1 inhibition. The substantial increase in the amount of RNA per cell induced by TCR activation was reduced by 50% by deficiency in mTORC1, but not in mTORC2 or in S6 kinases 1 and 2, which are activated downstream of mTORC1. RNA-seq data showed that mTORC1 deficiency reduced the abundance of all RNA biotypes, although rRNA processing was largely intact in activated T cells. Imaging cytometry with FISH probes for nascent pre-rRNA revealed that deletion of mTORC1, but not that of mTORC2, reduced the number and expansion of nucleolar sites of active transcription. Protein translation was consequently decreased by 50% in the absence of mTORC1. Inhibiting RNA polymerase I blocked not only proliferation but also mTORC1 signaling. Our data show that TCR signaling, mTORC1 activity, and ribosomal biosynthesis in the nucleolus regulate each other during biomass production in clonally expanding T cells.
Collapse
Affiliation(s)
- Teresa Rosenlehner
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Stefanie Pennavaria
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Batuhan Akçabozan
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Shiva Jahani
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Thomas J O'Neill
- Research Unit Signaling and Translation, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Tobias Straub
- Bioinformatics Core Facility, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Jan Kranich
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Reinhard Obst
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
27
|
Hamid MA, Pammer LM, Lentner TK, Doleschal B, Gruber R, Kocher F, Gasser E, Jöbstl A, Seeber A, Amann A. Immunotherapy for Microsatellite-Stable Metastatic Colorectal Cancer: Can we close the Gap between Potential and Practice? Curr Oncol Rep 2024; 26:1258-1270. [PMID: 39080202 PMCID: PMC11480176 DOI: 10.1007/s11912-024-01583-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 10/17/2024]
Abstract
PURPOSE OF REVIEW This review will explore various strategies to rendering MSS mCRCs susceptible to ICI. Moreover, we will provide an overview of potential biomarkers that may aid to better patient selection, and discuss ongoing efforts in this area of research. RECENT FINDINGS Colorectal cancer (CRC) ranks among the top three most common cancers worldwide. While significant advances in treatment strategies have improved the prognosis for patients in the early stages of the disease, treatment options for metastatic CRC (mCRC) remain limited. Although immune checkpoint inhibitors (ICI) have revolutionized the treatment of several malignancies, its efficacy in mCRC is largely confined to patients exhibiting a high microsatellite instability status (MSI-H). However, the vast majority of mCRC patients do not exhibit a MSI-H, but are microsatellite stable (MSS). In these patients ICIs are largely ineffective. So far, ICIs do not play a crucial role in patients with MSS mCRC, despite the promising data for inducing long-term remissions in other tumour entities. For this reason, novel treatment strategies are needed to overcome the primary resistance upon ICI in patients with MSS.
Collapse
Affiliation(s)
- Marwa Abdel Hamid
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Lorenz M Pammer
- Department of Gastroenterology and Hepatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Theresa K Lentner
- Clinical Department for Internal Medicine, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Bernhard Doleschal
- Department of Internal Medicine I for Hematology With Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Rebecca Gruber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Florian Kocher
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Elisabeth Gasser
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Jöbstl
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Arno Amann
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
28
|
Sigaud R, Brummer T, Kocher D, Milde T, Selt F. MOST wanted: navigating the MAPK-OIS-SASP-tumor microenvironment axis in primary pediatric low-grade glioma and preclinical models. Childs Nerv Syst 2024; 40:3209-3221. [PMID: 38789691 PMCID: PMC11511703 DOI: 10.1007/s00381-024-06463-z] [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: 03/28/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
Understanding the molecular and cellular mechanisms driving pediatric low-grade glioma (pLGG)-the most prevalent brain tumor in children-is essential for the identification and evaluation of novel effective treatments. This review explores the intricate relationship between the mitogen-activated protein kinase (MAPK) pathway, oncogene-induced senescence (OIS), the senescence-associated secretory phenotype (SASP), and the tumor microenvironment (TME), integrating these elements into a unified framework termed the MAPK/OIS/SASP/TME (MOST) axis. This integrated approach seeks to deepen our understanding of pLGG and improve therapeutic interventions by examining the MOST axis' critical influence on tumor biology and response to treatment. In this review, we assess the axis' capacity to integrate various biological processes, highlighting new targets for pLGG treatment, and the need for characterized in vitro and in vivo preclinical models recapitulating pLGG's complexity to test targets. The review underscores the need for a comprehensive strategy in pLGG research, positioning the MOST axis as a pivotal approach in understanding pLGG. This comprehensive framework will open promising avenues for patient care and guide future research towards inventive treatment options.
Collapse
Affiliation(s)
- Romain Sigaud
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| | - Tilman Brummer
- Institute, of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Biological Signaling Studies BIOSS, University of Freiburg and German Consortium for Translational Cancer Research (DKTK), Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Kocher
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Selt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
29
|
Lakhani NJ, Burris H, Miller WH, Huang M, Chen LC, Siu LL. A phase 1b study of the ERK inhibitor MK-8353 plus pembrolizumab in patients with advanced solid tumors. Invest New Drugs 2024; 42:581-589. [PMID: 39276176 PMCID: PMC11625062 DOI: 10.1007/s10637-024-01461-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/18/2024] [Indexed: 09/16/2024]
Abstract
Combining a checkpoint inhibitor with an inhibitor of extracellular signal-regulated kinase (ERK) may result in synergistic antitumor activity. We evaluated MK-8353, an ERK1 and ERK2 inhibitor, plus pembrolizumab in a phase 1b study in patients with advanced solid tumors. This open-label, nonrandomized, dose-escalation study (NCT02972034) enrolled adults with advanced solid tumors previously treated with 1‒5 prior lines of therapy. MK-8353 was administered orally in combination with pembrolizumab 200 mg every 3 weeks as follows: twice daily (arm A; MK-8353 50‒350 mg), once daily (arm B; MK-8353 50‒600 mg), or once daily every other week (arm C; MK-8353 50‒300 mg). The primary objective was evaluation of safety via occurrence of dose-limiting toxicities (DLTs). A secondary objective was objective response by RECIST v1.1 per investigator assessment. Among 110 evaluable patients (arm A, n = 22; arm B, n = 50; arm C, n = 38), median age was 58.0 (range, 35‒79) years and 50% had received 1 or 2 prior lines of therapy. DLTs occurred in 19 patients (n = 6 [27%], n = 8 [16%], and n = 5 [13%], respectively); the most frequent was grade 3 maculopapular rash (n = 15). Grade 3/4 treatment-related AEs occurred in 35% of patients; the most common were maculopapular rash (13%) and increased lipase (5%); none were grade 5. Eight patients (7%) attained an objective response (arm B, n = 7 [complete response, n = 1; partial response, n = 6]; arm C, n = 1 [complete response]). In conclusion, MK-8353 once daily plus pembrolizumab could be administered with a manageable toxicity profile but had modest antitumor activity in patients with advanced solid tumors.
Collapse
Affiliation(s)
| | | | - Wilson H Miller
- Department of Medicine and Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Mo Huang
- Merck & Co., Inc., Rahway, NJ, USA
| | | | - Lillian L Siu
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
30
|
Yu J, Xiang Y, Gao Y, Chang S, Kong R, Lv X, Yu J, Jin Y, Li C, Ma Y, Wang Z, Zhou J, Yuan H, Shang S, Hua F, Zhang X, Cui B, Li P. PKC α inhibitors promote breast cancer immune evasion by maintaining PD-L1 stability. Acta Pharm Sin B 2024; 14:4378-4395. [PMID: 39525583 PMCID: PMC11544271 DOI: 10.1016/j.apsb.2024.08.003] [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: 03/01/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 11/16/2024] Open
Abstract
Protein kinase C α (PKCα) regulates diverse biological functions of cancer cells and is a promising therapeutic target. However, clinical trials of PKC-targeted therapies have not yielded satisfactory results. Recent studies have also indicated a tumor-suppressive role of PKCs via unclear molecular mechanisms. In this study, we found that PKCα inhibition enhances CD8+ T-cell-mediated tumor evasion and abolishes antitumor activity in immunocompetent mice. We further identified PKCα as a critical regulator of programmed cell death-ligand 1 (PD-L1) and found that it enhances T-cell-dependent antitumor immunity in breast cancer by interacting with PD-L1 and suppressing PD-L1 expression. We demonstrated that PKCα-mediated PD-L1 phosphorylation promotes PD-L1 degradation through β transducin repeat-containing protein. Notably, the efficacy of PKCα inhibitors was intensified by synergizing with anti-PD-L1 mAb therapy to boost antitumor T-cell immunity in vivo. Clinical analysis revealed that PKCα expression is positively correlated with T-cell function and the interferon-gamma signature in patients with breast cancer. This study demonstrated the antitumor capability of PKCα, identified potential therapeutic strategies to avoid tumor evasion via PKC-targeted therapies, and provided a proof of concept for targeting PKCα in combination with anti-PD-L1 mAb therapy as a potential therapeutic approach against breast cancer, especially TNBC.
Collapse
Affiliation(s)
- Jiaojiao Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yujin Xiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuzhen Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shan Chang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Ren Kong
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Xiaoxi Lv
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jinmei Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yunjie Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chenxi Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yiran Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zhenhe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jichao Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongyu Yuan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuang Shang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Fang Hua
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiaowei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bing Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
31
|
Dong L, Choi H, Budhu S, Schulze I, Verma S, Mangarin LM, Estrada Nevarro V, Mehanna N, Khan JF, Venkatesh D, Thach D, Rosen N, Wolchok JD, Merghoub T. Intermittent MEK Inhibition with GITR Costimulation Rescues T-cell Function for Increased Efficacy with CTLA-4 Blockade in Solid Tumor Models. Cancer Immunol Res 2024; 12:1392-1408. [PMID: 38885362 DOI: 10.1158/2326-6066.cir-23-0729] [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: 09/12/2023] [Revised: 03/14/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
MEK inhibitors (MEKi) have shown limited success as a treatment for MAPK/ERK pathway-dependent cancers due to various resistance mechanisms tumor cells can employ. CH5126766 (CKI27) is an inhibitor that binds to MEK and prevents release of RAF, reducing the relief of negative feedback commonly observed with other MEKis. We observed that CKI27 increased MHC expression in tumor cells and improved T cell-mediated killing. Yet, CKI27 also decreased T-cell proliferation, activation, and cytolytic activity by inhibiting the MAPK/ERK pathway that is activated downstream of T-cell receptor signaling. Therefore, we aimed to balance the positive and negative immunomodulatory effects of MEKis for optimal combination with immunotherapy. Intermittent administration of CKI27 allowed T cells to partially recover and costimulation via GITR and OX-40 agonist antibodies completely alleviated inhibition of function. In Kras mutant lung and colon tumor mouse models, intermittent CKI27 and anti-GITR significantly decreased tumor growth and prolonged survival when further combined with CTLA-4 immune checkpoint blockade. Moreover, this triple combination increased CD8+ and CD4+ T-cell proliferation, activation, and effector/memory subsets in the tumor-draining lymph nodes and tumors and led to intratumoral regulatory T-cell destabilization. These data, collectively, will allow for more informed decisions when optimizing combination regimens by overcoming resistance, reducing toxicity, and generating long-term immune responses.
Collapse
Affiliation(s)
- Lauren Dong
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Hyejin Choi
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Sadna Budhu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Isabell Schulze
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Svena Verma
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Levi M Mangarin
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Valeria Estrada Nevarro
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Nezar Mehanna
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Jonathan F Khan
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Divya Venkatesh
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Daniel Thach
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Neal Rosen
- Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jedd D Wolchok
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Taha Merghoub
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
- Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, New York
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, New York
| |
Collapse
|
32
|
Aruquipa MPS, Donadio MS, Peixoto RD. Liver metastasis and resistance to immunotherapy in microsatellite stable colorectal cancer. A literature review. Ecancermedicalscience 2024; 18:1771. [PMID: 39430087 PMCID: PMC11489097 DOI: 10.3332/ecancer.2024.1771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Indexed: 10/22/2024] Open
Abstract
Background Microsatellite stable (MSS) metastatic colorectal cancer (CRC) remains predominantly managed with chemotherapy. The use of immunotherapy, whether alone or in combination with other systemic or local treatments, displays limited success, especially in the context of active liver metastases (LM). The mechanisms responsible for this resistance are not fully understood. Methods We conducted a comprehensive search across electronic databases such as Medline, PubMed, Google Scholar and ScienceDirect. This search targeted translational studies evaluating the liver tumour immune microenvironment and immune tolerance mechanisms in CRC with LM and prospective studies that assessed immunotherapy either as a standalone treatment or in combination with other systemic or local therapies for patients diagnosed with MSS CRC. Our primary objectives included elucidating the mechanisms of resistance originating from LM in a non-systematic literature review and presenting a summary of the outcomes observed in prospective trials utilising immune checkpoint inhibitors (ICIs), with a focus on the presence of LM. Findings There were 16 prospective trials evaluating immunotherapy for metastatic CRC comprising 1,713 patients. Response rates to immunotherapy inpatients with colorectal liver metastases (CRLM) varied from 0% to 23%. Overall, reduced or null responses to immunotherapy in the presence of liver metastasis in comparison to patients without liver involvement were observed. Conclusion Studies consistently show the resistance derived from classical ICI, both alone and in combination with other systemic treatments in patients with CRLM. The design of upcoming trials using immunotherapy should consider LM as a stratification factor or contemplate excluding patients with liver involvement.
Collapse
Affiliation(s)
| | - Mauro S Donadio
- Gastrointestinal Oncology Department, Oncoclinicas, São Paulo 04513-100, Brazil
- https://orcid.org/0000-0002-4705-4802
| | - Renata D Peixoto
- BC Cancer Agency, Vancouver, BC V5Z 4E6, Canada
- https://orcid.org/0000-0003-0053-7951
| |
Collapse
|
33
|
Gong J, Guo Y, Zhang Y, Ba Y, Chen T, Li W, Zhou C, Wang M, Yang H, Zhou Y, Cai Q, Wang Z, Huang G, Zhang W, Su R, Cai Z, Yue Z, Dou J, Li P, Wu R, Tse AN, Shen L. A Phase 1a/1b Dose Escalation/Expansion Study of the Anti-PD-1 Monoclonal Antibody Nofazinlimab in Chinese Patients with Solid Tumors or Lymphoma. Target Oncol 2024; 19:723-733. [PMID: 39231855 DOI: 10.1007/s11523-024-01091-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Immune checkpoint blockade with anti-programmed cell death 1 (PD-1) antibodies has demonstrated efficacy in multiple tumor types. Nofazinlimab is a humanized rat antibody targeting PD-1. A first-in-human study of nofazinlimab conducted in Australia found no dose-limiting toxicities (DLTs) and the maximum tolerated dose (MTD) was not reached in the range of 1-10 mg/kg. OBJECTIVE We evaluated nofazinlimab for multiple advanced malignancies in Chinese patients. PATIENTS AND METHODS This was a phase 1a/1b, open-label, multicenter, dose-escalation/expansion trial. In phase 1a, patients received an abbreviated dose escalation of nofazinlimab at 60 mg and 200 mg every 3 weeks (Q3W) to determine DLTs and the recommended phase 2 dose (RP2D). In phase 1b, patients received the RP2D (monotherapy/combination) in six arms by tumor type; DLTs were evaluated for nofazinlimab plus lenvatinib in the unresectable hepatocellular carcinoma (uHCC) arm. Safety (continuously monitored in patients who received nofazinlimab) and efficacy (patients with measurable baseline disease) were assessed. RESULTS Overall, 107 patients were eligible and received nofazinlimab. In phase 1a, no DLTs were observed; the RP2D was 200mg Q3W. In phase 1b, no DLTs were observed with nofazinlimab plus lenvatinib. The safety profile was consistent with that observed in the first-in-human study (NCT03475251). In phase 1b, 21/88 (23.9%) patients achieved confirmed objective responses, 26 (29.5%) had stable disease, and 9/20 (45.0%) patients with uHCC achieved confirmed objective responses to nofazinlimab plus lenvatinib. CONCLUSIONS Nofazinlimab was well tolerated in Chinese patients. Preliminary efficacy was encouraging, particularly for nofazinlimab plus lenvatinib in uHCC, which is being studied in an ongoing phase 3 trial. CLINICAL TRIAL REGISTRATION NCT03809767; registered 18 January 2019.
Collapse
Affiliation(s)
- Jifang Gong
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Ye Guo
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanqiao Zhang
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yi Ba
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tong Chen
- Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- The First Hospital of Jilin University, Changchun, Jilin, China
| | - Caicun Zhou
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mengzhao Wang
- Peking Union Medical College Hospital, Beijing, China
| | | | | | | | - Ziping Wang
- Peking University Cancer Hospital & Institute, Beijing, China
| | | | - Wei Zhang
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rila Su
- CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Zhongheng Cai
- CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Zenglian Yue
- CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Jinzhou Dou
- CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Peiqi Li
- CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Rachel Wu
- CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Archie N Tse
- CStone Pharmaceuticals (Suzhou) Co., Ltd., Suzhou, China
| | - Lin Shen
- Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
34
|
Lara-Vega I. Upgrading Melanoma Treatment: Promising Immunotherapies Combinations
in the Preclinical Mouse Model. CURRENT CANCER THERAPY REVIEWS 2024; 20:489-509. [DOI: 10.2174/0115733947263244231002042219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/27/2023] [Accepted: 08/25/2023] [Indexed: 01/03/2025]
Abstract
Background:
Melanoma, known for its high metastatic potential, does not respond well to
existing treatments in advanced stages. As a solution, immunotherapy-based treatments, including
anti-PD-1/L1 and anti-CTLA-4, have been developed and evaluated in preclinical mouse models to
overcome resistance. Although these treatments display the potential to suppress tumor growth, there
remains a crucial requirement for a thorough assessment of long-term efficacy in preventing metastasis
or recurrence and improving survival rates.
Methods:
From 2016 onwards, a thorough examination of combined immunotherapies for the treatment
of cutaneous melanoma in preclinical mouse models was conducted. The search was conducted
using MeSH Terms algorithms in PubMed®, resulting in the identification of forty-five studies that
met the rigorous inclusion criteria for screening.
Results:
The C57 mouse model bearing B16-melanoma has been widely utilized to assess the efficacy
of immunotherapies. The combination of therapies has demonstrated a synergistic impact, leading
to potent antitumor activity. One extensively studied method for establishing metastatic models involves
the intravenous administration of malignant cells, with several combined therapies under investigation.
The primary focus of evaluation has been on combined immunotherapies utilizing PD-
1/L1 and CTLA-4 blockade, although alternative immunotherapies not involving PD-1/L1 and
CTLA-4 blockade have also been identified. Additionally, the review provides detailed treatment regimens
for each combined approach.
Conclusion:
The identification of techniques for generating simulated models of metastatic melanoma
and investigating various therapeutic combinations will greatly aid in evaluating the overall systemic
efficacy of immunotherapy. This will be especially valuable for conducting short-term preclinical
experiments that have the potential for clinical studies.
Collapse
Affiliation(s)
- Israel Lara-Vega
- National School of Biological Sciences, IPN. Av. Wilfrido Massieu s/n, Professional Unit Adolfo Lopez Mateos, Mexico
City, CP 07738, Mexico
| |
Collapse
|
35
|
Piercey O, Tie J, Hollande F, Wong HL, Mariadason J, Desai J. BRAF V600E-Mutant Metastatic Colorectal Cancer: Current Evidence, Future Directions, and Research Priorities. Clin Colorectal Cancer 2024; 23:215-229. [PMID: 38816264 DOI: 10.1016/j.clcc.2024.04.004] [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: 04/03/2024] [Accepted: 04/24/2024] [Indexed: 06/01/2024]
Abstract
BRAFV600E-mutant metastatic colorectal cancer represents a distinct molecular phenotype known for its aggressive biological behavior, resistance to standard therapies, and poor survival rates. Improved understanding of the biology of the BRAF oncogene has led to the development of targeted therapies that have paved the way for a paradigm shift in managing this disease. However, despite significant recent advancements, responses to targeted therapies are short-lived, and several challenges remain. In this review, we discuss how progress in treating BRAFV600E-mutant metastatic colorectal cancer has been made through a better understanding of its unique biological and clinical features. We provide an overview of the evidence to support current treatment approaches and discuss critical areas of need and future research strategies that hold the potential to refine clinical practice further. We also discuss some challenging aspects of managing this disease, particularly the complexity of acquired resistance mechanisms that develop under the selective pressure of targeted therapies and rational strategies being investigated to overcome them.
Collapse
Affiliation(s)
- Oliver Piercey
- Peter MacCallum Cancer Centre, Melbourne, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, Australia; Department of Clinical Pathology, The University of Melbourne, Australia.
| | - Jeanne Tie
- Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Frederic Hollande
- Centre for Cancer Research, The University of Melbourne, Melbourne, Australia; Department of Clinical Pathology, The University of Melbourne, Australia
| | - Hui-Li Wong
- Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - John Mariadason
- Olivia Newton John Cancer Wellness and Research Centre, Heidelberg, Australia; School of Medicine, La Trobe University, Melbourne, Australia
| | - Jayesh Desai
- Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
36
|
Chibaya L, DeMarco KD, Lusi CF, Kane GI, Brassil ML, Parikh CN, Murphy KC, Chowdury SR, Li J, Ma B, Naylor TE, Cerrutti J, Mori H, Diaz-Infante M, Peura J, Pitarresi JR, Zhu LJ, Fitzgerald KA, Atukorale PU, Ruscetti M. Nanoparticle delivery of innate immune agonists combined with senescence-inducing agents promotes T cell control of pancreatic cancer. Sci Transl Med 2024; 16:eadj9366. [PMID: 39196958 PMCID: PMC11811823 DOI: 10.1126/scitranslmed.adj9366] [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: 08/04/2023] [Revised: 03/18/2024] [Accepted: 06/11/2024] [Indexed: 08/30/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has quickly risen to become the third leading cause of cancer-related death in the United States. This is in part because of its fibrotic tumor microenvironment (TME) that contributes to poor vascularization and immune infiltration and subsequent chemo- and immunotherapy failure. Here, we investigated an immunotherapy approach combining delivery of stimulator of interferon genes (STING) and Toll-like receptor 4 (TLR4) innate immune agonists by lipid-based nanoparticle (NP) coencapsulation with senescence-inducing RAS-targeted therapies, which can remodel the immune suppressive PDAC TME through the senescence-associated secretory phenotype. Treatment of transplanted and autochthonous PDAC mouse models with these regimens led to enhanced uptake of NPs by multiple cell types in the PDAC TME, induction of type I interferon and other proinflammatory signaling pathways, increased antigen presentation by tumor cells and antigen-presenting cells, and subsequent activation of both innate and adaptive immune responses. This two-pronged approach produced potent T cell-driven and type I interferon-mediated tumor regression and long-term survival in preclinical PDAC models dependent on both tumor and host STING activation. STING and TLR4-mediated type I interferon signaling was also associated with enhanced natural killer and CD8+ T cell immunity in human PDAC samples. Thus, combining localized immune agonist delivery with systemic tumor-targeted therapy can orchestrate a coordinated type I interferon-driven innate and adaptive immune response with durable antitumor efficacy against PDAC.
Collapse
Affiliation(s)
- Loretah Chibaya
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Kelly D. DeMarco
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Christina F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Griffin I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Meghan L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Chaitanya N. Parikh
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Katherine C. Murphy
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Shreya R. Chowdury
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Junhui Li
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Boyang Ma
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Tiana E. Naylor
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Julia Cerrutti
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Haruka Mori
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Miranda Diaz-Infante
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Jessica Peura
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Jason R. Pitarresi
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Katherine A. Fitzgerald
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Prabhani U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Immunology and Microbiology Program, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| |
Collapse
|
37
|
Ge J, Ge J, Tang G, Xiong D, Zhu D, Ding X, Zhou X, Sang M. Machine learning-based identification of biomarkers and drugs in immunologically cold and hot pancreatic adenocarcinomas. J Transl Med 2024; 22:775. [PMID: 39152432 PMCID: PMC11328457 DOI: 10.1186/s12967-024-05590-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Pancreatic adenocarcinomas (PAADs) often exhibit a "cold" or immunosuppressive tumor milieu, which is associated with resistance to immune checkpoint blockade therapy; however, the underlying mechanisms are incompletely understood. Here, we aimed to improve our understanding of the molecular mechanisms occurring in the tumor microenvironment and to identify biomarkers, therapeutic targets, and potential drugs to improve PAAD treatment. METHODS Patients were categorized according to immunologically hot or cold PAAD subtypes with distinct disease outcomes. Cox regression and weighted correlation network analysis were performed to construct a novel gene signature, referred to as 'Downregulated in hot tumors, Prognostic, and Immune-Related Genes' (DPIRGs), which was used to develop prognostic models for PAAD via machine learning (ML). The role of DPIRGs in PAAD was comprehensively analyzed, and biomarker genes able to distinguish PAAD immune subtypes and predict prognosis were identified by ML. The expression of biomarkers was verified using public single-cell transcriptomic and proteomic resources. Drug candidates for turning cold tumors hot and corresponding target proteins were identified via molecular docking studies. RESULTS Using the DPIRG signature as input data, a combination of survival random forest and partial least squares regression Cox was selected from 137 ML combinations to construct an optimized PAAD prognostic model. The effects and molecular mechanisms of DPIRGs were investigated by analysis of genetic/epigenetic alterations, immune infiltration, pathway enrichment, and miRNA regulation. Biomarkers and potential therapeutic targets, including PLEC, TRPV1, and ITGB4, among others, were identified, and the cell type-specific expression of the biomarkers was validated. Drug candidates, including thalidomide, SB-431542, and bleomycin A2, were identified based on their ability to modulate DPIRG expression favorably. CONCLUSIONS By combining multiple ML algorithms, we developed a novel prognostic model with excellent performance in PAAD cohorts. ML also proved to be powerful for identifying biomarkers and potential targets for improved PAAD patient stratification and immunotherapy.
Collapse
Affiliation(s)
- Jia Ge
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226001, China
| | - Juan Ge
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226001, China
- Department of Respiratory Medicine, Affiliated Nantong Hospital of Shanghai University, Nantong, 226011, China
| | - Gu Tang
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226001, China
| | - Dejun Xiong
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226001, China
| | - Dongyan Zhu
- Department of Rehabilitation, the Second Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Xiaoling Ding
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Mengmeng Sang
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
38
|
Steiner C, Denlinger N, Huang X, Yang Y. Stem-like CD8 + T cells in cancer. Front Immunol 2024; 15:1426418. [PMID: 39211052 PMCID: PMC11357971 DOI: 10.3389/fimmu.2024.1426418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Stem-like CD8+ T cells (TSL) are a subset of immune cells with superior persistence and antitumor immunity. They are TCF1+ PD-1+ and important for the expansion of tumor specific CD8+ T cells in response to checkpoint blockade immunotherapy. In acute infections, naïve CD8+ T cells differentiate into effector and memory CD8+ T cells; in cancer and chronic infections, persistent antigen stimulation can lead to T cell exhaustion. Recent studies have highlighted the dichotomy between late dysfunctional (or exhausted) T cells (TLD) that are TCF1- PD-1+ and self-renewing TCF1+ PD-1+ TSL from which they derive. TCF1+ TSL cells are considered to have stem cell-like properties akin to memory T cell populations and can give rise to cytotoxic effector and transitory T cell phenotypes (TTE) which mediate tumor control. In this review, we will discuss recent advances made in research on the formation and expansion of TSL, as well as distinct niches required for their differentiation and maintenance in the setting of cancer. We will also discuss potential strategies to generate these cells, with clinical implications for stemness enhancement in vaccine design, immune checkpoint blockade (ICB), and adoptive T cell therapies.
Collapse
Affiliation(s)
| | | | - Xiaopei Huang
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Yiping Yang
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
39
|
Liu W, Zhou H, Lai W, Hu C, Xu R, Gu P, Luo M, Zhang R, Li G. The immunosuppressive landscape in tumor microenvironment. Immunol Res 2024; 72:566-582. [PMID: 38691319 DOI: 10.1007/s12026-024-09483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Recent advances in cancer immunotherapy, especially immune checkpoint inhibitors (ICIs), have revolutionized the clinical outcome of many cancer patients. Despite the fact that impressive progress has been made in recent decades, the response rate remains unsatisfactory, and many patients do not benefit from ICIs. Herein, we summarized advanced studies and the latest insights on immune inhibitory factors in the tumor microenvironment. Our in-depth discussion and updated landscape of tumor immunosuppressive microenvironment may provide new strategies for reversing tumor immune evasion, enhancing the efficacy of ICIs therapy, and ultimately achieving a better clinical outcome.
Collapse
Affiliation(s)
- Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Huyue Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rufu Xu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Peng Gu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Menglin Luo
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| |
Collapse
|
40
|
Schnell A. Stem-like T cells in cancer and autoimmunity. Immunol Rev 2024; 325:9-22. [PMID: 38804499 DOI: 10.1111/imr.13356] [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] [Indexed: 05/29/2024]
Abstract
Stem-like T cells are characterized by their ability to self-renew, survive long-term, and give rise to a heterogeneous pool of effector and memory T cells. Recent advances in single-cell RNA-sequencing (scRNA-seq) and lineage tracing technologies revealed an important role for stem-like T cells in both autoimmunity and cancer. In cancer, stem-like T cells constitute an important arm of the anti-tumor immune response by giving rise to effector T cells that mediate tumor control. In contrast, in autoimmunity stem-like T cells perform an unfavorable role by forming a reservoir of long-lived autoreactive cells that replenish the pathogenic, effector T-cell pool and thereby driving disease pathology. This review provides background on the discovery of stem-like T cells and their function in cancer and autoimmunity. Moreover, the influence of the microbiota and metabolism on the stem-like T-cell pool is summarized. Lastly, the implications of our knowledge about stem-like T cells for clinical treatment strategies for cancer and autoimmunity will be discussed.
Collapse
Affiliation(s)
- Alexandra Schnell
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
41
|
Savage WM, Yeary MD, Tang AJ, Sperring CP, Argenziano MG, Adapa AR, Yoh N, Canoll P, Bruce JN. Biomarkers of immunotherapy in glioblastoma. Neurooncol Pract 2024; 11:383-394. [PMID: 39006524 PMCID: PMC11241363 DOI: 10.1093/nop/npae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Glioblastoma (GBM) is the most common primary brain cancer, comprising half of all malignant brain tumors. Patients with GBM have a poor prognosis, with a median survival of 14-15 months. Current therapies for GBM, including chemotherapy, radiotherapy, and surgical resection, remain inadequate. Novel therapies are required to extend patient survival. Although immunotherapy has shown promise in other cancers, including melanoma and non-small lung cancer, its efficacy in GBM has been limited to subsets of patients. Identifying biomarkers of immunotherapy response in GBM could help stratify patients, identify new therapeutic targets, and develop more effective treatments. This article reviews existing and emerging biomarkers of clinical response to immunotherapy in GBM. The scope of this review includes immune checkpoint inhibitor and antitumoral vaccination approaches, summarizing the variety of molecular, cellular, and computational methodologies that have been explored in the setting of anti-GBM immunotherapies.
Collapse
Affiliation(s)
- William M Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Mitchell D Yeary
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Anthony J Tang
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Colin P Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Arjun R Adapa
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Nina Yoh
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
42
|
Yang C, Zhao L, Lin Y, Wang S, Ye Y, Shen Z. Improving the efficiency of immune checkpoint inhibitors for metastatic pMMR/MSS colorectal cancer: Options and strategies. Crit Rev Oncol Hematol 2024; 200:104204. [PMID: 37984588 DOI: 10.1016/j.critrevonc.2023.104204] [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: 07/13/2023] [Revised: 10/24/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment and been extensively used for patients with metastastic colorectal cancer (mCRC), especially those harboring deficient mismatch repair/ microsatellite instability (dMMR/MSI). However, the majority of mCRC are classified as proficient mismatch repair/microsatellite stability(pMMR/MSS) type characterized by a cold immune microenvironment, rendering them generally unresponsive to ICIs. How to improve the efficacy of ICIs for these patients is an important issue to be solved. On the one hand, it is urgent to discover the predictive biomarkers and clinical characteristics associated with effectiveness and expand the subset of pMMR/MSS mCRC patients who benefit from ICIs. Additionally, combined strategies are being explored to modulate the immune microenvironment of pMMR/MSS CRC and facilitate the conversion of cold tumors into hot tumors. In this review, we have focused on the recent advancements in the predictive biomarkers and combination therapeutic strategies with ICIs for pMMR/MSS mCRC.
Collapse
Affiliation(s)
- Changjiang Yang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Long Zhao
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Yilin Lin
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Shan Wang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China.
| |
Collapse
|
43
|
Bai W, Yang L, Qiu J, Zhu Z, Wang S, Li P, Zhou D, Wang H, Liao Y, Yu Y, Yang Z, Wen P, Zhang D. Single-cell analysis of CD4+ tissue residency memory cells (TRMs) in adult atopic dermatitis: A new potential mechanism. Genomics 2024; 116:110870. [PMID: 38821220 DOI: 10.1016/j.ygeno.2024.110870] [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: 07/23/2023] [Revised: 05/09/2024] [Accepted: 05/25/2024] [Indexed: 06/02/2024]
Abstract
The pathophysiology of atopic dermatitis (AD) is complex. CD4+ T cells play an essential role in the development of lesions in AD. However, the underlying mechanism remains unclear. In the present study, we investigated the differentially expressed genes (DEGs) between adult AD lesioned and non-lesioned skin using two datasets from the Gene Expression Omnibus (GEO) database. 62 DEGs were shown to be related to cytokine response. Compared to non-lesioned skin, lesioned skin showed immune infiltration with increased numbers of activated natural killer (NK) cells and CD4+ T memory cells (p < 0.01). We then identified 13 hub genes with a strong association with CD4+ T cells using weighted correlation network analysis. Single-cell analysis of AD detected a novel CD4+ T subcluster, CD4+ tissue residency memory cells (TRMs), which were verified through immunohistochemistry (IHC) to be increased in the dermal area of AD. The significant relationship between CD4+ TRM and AD was assessed through further analyses. FOXO1 and SBNO2, two of the 13 hub genes, were characteristically expressed in the CD4+ TRM, but down-regulated in IFN-γ/TNF-α-induced HaCaT cells, as shown using quantitative polymerase chain reaction (qPCR). Moreover, SBNO2 expression was associated with increased Th1 infiltration in AD (p < 0.05). In addition, genes filtered using Mendelian randomization were positively correlated with CD4+ TRM and were highly expressed in IFN-γ/TNF-α-induced HaCaT cells, as determined using qPCR and western blotting. Collectively, our results revealed that the newly identified CD4+ TRM may be involved in the pathogenesis of adult AD.
Collapse
Affiliation(s)
- Wenxuan Bai
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Le Yang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jing Qiu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zihan Zhu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shuxing Wang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Peidi Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Dawei Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hongyi Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxuan Liao
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yao Yu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zijiang Yang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Puqiao Wen
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Di Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
44
|
Singh M, Morris VK, Bandey IN, Hong DS, Kopetz S. Advancements in combining targeted therapy and immunotherapy for colorectal cancer. Trends Cancer 2024; 10:598-609. [PMID: 38821852 DOI: 10.1016/j.trecan.2024.05.001] [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: 01/11/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/02/2024]
Abstract
Colorectal cancer (CRC) is a prevalent gastrointestinal cancer posing significant clinical challenges. CRC management traditionally involves surgery, often coupled with chemotherapy. However, unresectable or metastatic CRC (mCRC) presents a complex challenge necessitating innovative treatment strategies. Targeted therapies have emerged as the cornerstone of treatment in such cases, with interventions tailored to specific molecular attributes. Concurrently, immunotherapies have revolutionized cancer treatment by harnessing the immune system to combat malignant cells. This review explores the evolving landscape of CRC treatment, focusing on the synergy between immunotherapies and targeted therapies, thereby offering new avenues for enhancing the effectiveness of therapy for CRC.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Van Karlyle Morris
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irfan N Bandey
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
45
|
Imyanitov EN, Mitiushkina NV, Kuligina ES, Tiurin VI, Venina AR. Pathways and targeting avenues of BRAF in non-small cell lung cancer. Expert Opin Ther Targets 2024; 28:613-622. [PMID: 38941191 DOI: 10.1080/14728222.2024.2374742] [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: 03/10/2024] [Accepted: 06/27/2024] [Indexed: 06/30/2024]
Abstract
INTRODUCTION BRAF is a serine-threonine kinase implicated in the regulation of MAPK signaling cascade. BRAF mutation-driven activation occurs in approximately 2-4% of treatment-naive non-small cell carcinomas (NSCLCs). BRAF upregulation is also often observed in tumors with acquired resistance to receptor tyrosine kinase inhibitors (TKIs). AREAS COVERED This review describes the spectrum of BRAF mutations and their functional roles, discusses treatment options available for BRAF p.V600 and non-V600 mutated NSCLCs, and identifies some gaps in the current knowledge. EXPERT OPINION Administration of combined BRAF/MEK inhibitors usually produces significant, although often a short-term, benefit to NSCLC patients with BRAF V600 (class 1) mutations. There are no established treatments for BRAF class 2 (L597, K601, G464, G469A/V/R/S, fusions, etc.) and class 3 (D594, G596, G466, etc.) mutants, which account for up to two-thirds of BRAF-driven NSCLCs. Many important issues related to the use of immune therapy for the management of BRAF-mutated NSCLC deserve further investigation. The rare occurrence of BRAF mutations in NSCLC is compensated by high overall incidence of lung cancer disease; therefore, clinical studies on BRAF-associated NSCLC are feasible.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, Russia
| | - Natalia V Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Ekatherina Sh Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Vladislav I Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Aigul R Venina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| |
Collapse
|
46
|
Norgard RJ, Budhani P, O'Brien SA, Xia Y, Egan JN, Flynn B, Tagore JR, Seco J, Peet GW, Mikucka A, Wasti R, Chan LC, Hinkel M, Martinez-Morilla S, Pignatelli J, Trapani F, Corse E, Feng D, Kostyrko K, Hofmann MH, Liu K, Kashyap AS. Reshaping the Tumor Microenvironment of KRASG12D Pancreatic Ductal Adenocarcinoma with Combined SOS1 and MEK Inhibition for Improved Immunotherapy Response. CANCER RESEARCH COMMUNICATIONS 2024; 4:1548-1560. [PMID: 38727236 PMCID: PMC11191876 DOI: 10.1158/2767-9764.crc-24-0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/23/2024]
Abstract
KRAS inhibitors have demonstrated exciting preclinical and clinical responses, although resistance occurs rapidly. Here, we investigate the effects of KRAS-targeting therapies on the tumor microenvironment using a library of KrasG12D, p53-mutant, murine pancreatic ductal adenocarcinoma-derived cell lines (KPCY) to leverage immune-oncology combination strategies for long-term tumor efficacy. Our findings show that SOS1 and MEK inhibitors (SOS1i+MEKi) suppressed tumor growth in syngeneic models and increased intratumoral CD8+ T cells without durable responses. Single-cell RNA sequencing revealed an increase in inflammatory cancer-associated fibroblasts (iCAF), M2 macrophages, and a decreased dendritic cell (DC) quality that ultimately resulted in a highly immunosuppressive microenvironment driven by IL6+ iCAFs. Agonist CD40 treatment was effective to revert macrophage polarization and overcome the lack of mature antigen-presenting DCs after SOS1i+MEKi therapy. Treatment increased the overall survival of KPCY tumor-bearing mice. The addition of checkpoint blockade to SOS1i+MEKi combination resulted in tumor-free mice with established immune memory. Our data suggest that KRAS inhibition affects myeloid cell maturation and highlights the need for combining KRAS cancer-targeted therapy with myeloid activation to enhance and prolong antitumor effects. SIGNIFICANCE Combination of SOS1 and MEK inhibitors increase T cell infiltration while blunting pro-immune myeloid cell maturation and highlights the need for combining KRAS cancer-targeted therapy with myeloid activation to enhance and prolong anti-tumor effects.
Collapse
Affiliation(s)
- Robert J. Norgard
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Pratha Budhani
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Sarah A. O'Brien
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Youli Xia
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Jessica N. Egan
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Brianna Flynn
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Joshua R. Tagore
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Joseph Seco
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Gregory W. Peet
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Ania Mikucka
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Ruby Wasti
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Li-Chuan Chan
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Melanie Hinkel
- Late Stage Cancer Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Sandra Martinez-Morilla
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Jeanine Pignatelli
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Francesca Trapani
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Emily Corse
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Di Feng
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Kaja Kostyrko
- Late Stage Cancer Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Marco H. Hofmann
- Cancer Pharmacology and Disease Positioning Department, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Kang Liu
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Abhishek S. Kashyap
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| |
Collapse
|
47
|
Bullock KK, Richmond A. Beyond Anti-PD-1/PD-L1: Improving Immune Checkpoint Inhibitor Responses in Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:2189. [PMID: 38927895 PMCID: PMC11201651 DOI: 10.3390/cancers16122189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
The introduction of anti-programmed cell death protein-1 (anti-PD-1) to the clinical management of triple-negative breast cancer (TNBC) represents a breakthrough for a disease whose treatment has long relied on the standards of chemotherapy and surgery. Nevertheless, few TNBC patients achieve a durable remission in response to anti-PD-1, and there is a need to develop strategies to maximize the potential benefit of immune checkpoint inhibition (ICI) for TNBC patients. In the present review, we discuss three conceptual strategies to improve ICI response rates in TNBC patients. The first effort involves improving patient selection. We discuss proposed biomarkers of response and resistance to anti-PD-1, concluding that an optimal biomarker will likely be multifaceted. The second effort involves identifying existing targeted therapies or chemotherapies that may synergize with ICI. In particular, we describe recent efforts to use inhibitors of the PI3K/AKT or RAS/MAPK/ERK pathways in combination with ICI. Third, considering the possibility that targeting the PD-1 axis is not the most promising strategy for TNBC treatment, we describe ongoing efforts to identify novel immunotherapy strategies.
Collapse
Affiliation(s)
| | - Ann Richmond
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA;
| |
Collapse
|
48
|
Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol 2024; 17:40. [PMID: 38835055 PMCID: PMC11151541 DOI: 10.1186/s13045-024-01561-6] [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: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Pineda JMB, Bradley RK. DUX4 is a common driver of immune evasion and immunotherapy failure in metastatic cancers. eLife 2024; 12:RP89017. [PMID: 38829686 PMCID: PMC11147511 DOI: 10.7554/elife.89017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Cancer immune evasion contributes to checkpoint immunotherapy failure in many patients with metastatic cancers. The embryonic transcription factor DUX4 was recently characterized as a suppressor of interferon-γ signaling and antigen presentation that is aberrantly expressed in a small subset of primary tumors. Here, we report that DUX4 expression is a common feature of metastatic tumors, with ~10-50% of advanced bladder, breast, kidney, prostate, and skin cancers expressing DUX4. DUX4 expression is significantly associated with immune cell exclusion and decreased objective response to PD-L1 blockade in a large cohort of urothelial carcinoma patients. DUX4 expression is a significant predictor of survival even after accounting for tumor mutational burden and other molecular and clinical features in this cohort, with DUX4 expression associated with a median reduction in survival of over 1 year. Our data motivate future attempts to develop DUX4 as a biomarker and therapeutic target for checkpoint immunotherapy resistance.
Collapse
Affiliation(s)
- Jose Mario Bello Pineda
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Basic Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Medical Scientist Training Program, University of WashingtonSeattleUnited States
| | - Robert K Bradley
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Basic Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| |
Collapse
|
50
|
Peng K, Liu Y, Liu S, Wang Z, Zhang H, He W, Jin Y, Wang L, Xia X, Xia L. Targeting MEK/COX-2 axis improve immunotherapy efficacy in dMMR colorectal cancer with PIK3CA overexpression. Cell Oncol (Dordr) 2024; 47:1043-1058. [PMID: 38315285 DOI: 10.1007/s13402-024-00916-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 02/07/2024] Open
Abstract
PURPOSE PIK3CA mutation or overexpression is associated with immunotherapy resistance in multiple cancer types, but is also paradoxically associated with benefit of COX-2 inhibition on patient survival of colorectal cancer (CRC) with mismatch repair deficiency (dMMR). This study examined whether and how PIK3CA status affected COX-2-mediated tumor inflammation and immunotherapy response of dMMR CRC. METHODS Murine colon cancer cells MC38, CT26, and CT26-Mlh1-KO were used to construct PIK3CA knockdown and overexpression models to mimic dMMR CRC with PIK3CA dysregulation, and xenograft models were used to evaluate how PIK3CA regulate COX-2 expression, CD8+ T cells infiltration, tumor growth, and therapy response to anti-PD-L1 treatment using immunocompetent mice. Western blot was carried out to delineate the signaling pathways in human and mouse cancer cells, and immunohistochemical analysis together with bioinformatics analysis using human patient samples. RESULTS PIK3CA upregulates COX-2 expression through MEK/ERK signaling pathway independent of AKT signaling to promote tumor inflammation and immunosuppression. PIK3CA knockdown profoundly reduced CT26 tumor growth in a CD8+ T cell-dependent manner, while PIK3CA overexpression significantly inhibited CD8+ T cells infiltration and promoted tumor growth. Furthermore, MEK or COX-2 inhibition augmented the anti-tumor activity of anti-PD-L1 immunotherapy on dMMR CRC mouse models, accompanied with increased CD8+ T cells infiltration and activated tumor microenvironment. CONCLUSION Our results identified that the PIK3CA hyperactivation in dMMR CRC upregulated COX-2 through MEK signaling, which inhibited CD8+ T cells infiltration and promoted tumor growth, together led to immunotherapy resistance. COX-2 or MEK inhibition may relieve therapy resistance and promote therapy efficacy of anti-PD-1/PD-L1 immunotherapy for treating dMMR CRC with PIK3CA overexpression or activating mutation.
Collapse
Affiliation(s)
- Kunwei Peng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Medical Oncology, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, China
| | - Yongxiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Shousheng Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zining Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Huanling Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Wenzhuo He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yanan Jin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lei Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.
| | - Liangping Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.
- VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|