1
|
Han RY, Gan LJ, Lang MR, Ren SH, Liu DM, Li GT, Liu YY, Tian XD, Zhu KW, Sun LY, Chen L, Song TQ. Lenvatinib, sintilimab combined interventional treatment vs bevacizumab, sintilimab combined interventional treatment for intermediate-advanced unresectable hepatocellular carcinoma. World J Gastroenterol 2024; 30:4620-4635. [DOI: 10.3748/wjg.v30.i43.4620] [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: 07/12/2024] [Revised: 09/21/2024] [Accepted: 10/21/2024] [Indexed: 10/31/2024] Open
Abstract
BACKGROUND Bevacizumab and sintilimab combined interventional treatment (BeSiIT) and L envatinib and sintilimab combined interventional treatment (LeSiIT) are two commonly used therapeutic regimens for intermediate-advanced hepatocellular carcinoma (HCC) in clinical practice.
AIM To compare the clinical efficacy and safety of BeSiIT and LeSiIT for the treatment of intermediate and advanced HCC.
METHODS Patients diagnosed with intermediate-advanced HCC and initially treated with BeSiIT or LeSiIT in the Tianjin Medical University Cancer Institute and Hospital between February 2020 and July 2021 were included. The primary endpoint was progression-free survival (PFS), and the secondary endpoints were overall survival (OS), objective response rate (ORR), disease control rate (DCR), conversion rate, and treatment-related adverse events.
RESULTS Total 127 patients met the inclusion criteria and were divided into BeSiIT and LeSiIT groups. Twenty-eight and fifty patients in the BeSiIT and LeSiIT groups, respectively, were assessed after 1:2 propensity score matching. PFS and OS rates were not significantly different between the two groups. No significant variations were noted in ORRs or DCRs according to the Response Evaluation Criteria in Solid Tumors (RECIST), and modified RECIST. BeSiIT group showed a better conversion rate than the LeSiIT group (P = 0.043). Both groups showed manageable toxicity profiles. Multivariate analysis showed that the independent factors associated with PFS were alpha-fetoprotein levels and carcinoembryonic antigen score.
CONCLUSION In intermediate-to-advanced HCC, the BeSiIT and LeSiIT groups exhibited acceptable toxicities and comparable PFS, OS, and ORR.
Collapse
Affiliation(s)
- Ru-Yu Han
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Lei-Juan Gan
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Meng-Ran Lang
- Department of Hepatobiliary Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shao-Hua Ren
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Dong-Ming Liu
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Guang-Tao Li
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Ya-Yue Liu
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Xin-Di Tian
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Kang-Wei Zhu
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Li-Yu Sun
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Lu Chen
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| | - Tian-Qiang Song
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin 300060, China
| |
Collapse
|
2
|
Abramson JS, Ku M, Hertzberg M, Huang HQ, Fox CP, Zhang H, Yoon DH, Kim WS, Abdulhaq H, Townsend W, Herbaux C, Zaucha JM, Zhang QY, Chang H, Liu Y, Cheah CY, Ghesquieres H, Simko S, Orellana-Noia V, Ta R, Relf J, Dixon M, Kallemeijn M, Mulvihill E, Huang H, Lundberg L, Gregory GP. Glofitamab plus gemcitabine and oxaliplatin (GemOx) versus rituximab-GemOx for relapsed or refractory diffuse large B-cell lymphoma (STARGLO): a global phase 3, randomised, open-label trial. Lancet 2024; 404:1940-1954. [PMID: 39550172 DOI: 10.1016/s0140-6736(24)01774-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Glofitamab monotherapy induces durable remission in patients with relapsed or refractory diffuse large B-cell lymphoma after two or more previous therapies, but has not previously been assessed as a second-line therapy. We investigated the efficacy and safety of glofitamab plus gemcitabine-oxaliplatin (Glofit-GemOx) versus rituximab (R)-GemOx in patients with relapsed or refractory diffuse large B-cell lymphoma. METHODS The phase 3, randomised, open-label STARGLO trial was done at 62 centres in 13 countries in Asia and Australia, Europe, and North America. We recruited transplant-ineligible patients (aged ≥18 years) with histologically confirmed relapsed or refractory diffuse large B-cell lymphoma after one or more previous therapies. Patients were randomly assigned in permuted blocks (block size of six) via an interactive voice or web response system (2:1; stratified by 1 vs ≥2 previous lines of therapy and relapsed vs refractory status) to Glofit-GemOx (intravenous gemcitabine 1000 mg/m2 and oxaliplatin 100 mg/m2 plus glofitamab step-up dosing to 30 mg; for a total of eight cycles, plus four additional cycles of glofitamab monotherapy) or R-GemOx (intravenous gemcitabine 1000 mg/m2 and oxaliplatin 100 mg/m2 plus rituximab 375 mg/m2; for a total of eight cycles). The trial independent review committee, which evaluated all response-based endpoints, was masked to treatment assignment. The primary endpoint was overall survival. Efficacy analyses were by intention to treat in all randomly assigned patients. We present results from both the primary analysis (cutoff: March 29, 2023) and updated analysis after all patients had completed study therapy (cutoff: Feb 16, 2024). Safety analyses included all patients who received any study treatment. This study is registered with ClinicalTrials.gov, NCT04408638, and is ongoing (closed to recruitment). FINDINGS From Feb 23, 2021, to March 14, 2023, 274 patients were enrolled and randomly assigned to receive Glofit-GemOx (n=183) or R-GemOx (n=91). 158 (58%) patients were male and 116 (42%) were female; median age was 68 years (IQR 58-74). At the primary analysis after a median follow-up of 11·3 months (95% CI 9·6-12·7), overall survival was significantly improved with Glofit-GemOx versus R-GemOx (median not estimable [NE; 95% CI 13·8 months-NE] vs 9·0 months [7·3-14·4]; hazard ratio [HR] 0·59 [95% CI 0·40-0·89]; p=0·011). At the updated analysis after a median follow-up of 20·7 months (19·9-23·3), a consistent improvement in overall survival was observed with Glofit-GemOx versus R-GemOx (median 25·5 months [18·3-NE] vs 12·9 months [7·9-18·5]; HR 0·62 [0·43-0·88]). In the safety sets, 180 (100%) patients in the Glofit-GemOx group and 84 (96%) of 88 patients in the R-GemOx group had at least one adverse event during the study period. Cytokine release syndrome occurred in 76 (44%) of 172 glofitamab-exposed patients and was predominantly low grade. Deaths related to glofitamab or rituximab occurred in five (3%) patients in the Glofit-GemOx group and in one (1%) patient in the R-GemOx group. INTERPRETATION Glofit-GemOx had a significant overall survival benefit compared with R-GemOx, supporting its use in transplant-ineligible patients with relapsed or refractory diffuse large B-cell lymphoma after one or more previous lines of therapy. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
| | - Matthew Ku
- St Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Mark Hertzberg
- Department of Haematology, Prince of Wales Hospital and UNSW, Sydney, NSW, Australia
| | | | | | - Huilai Zhang
- Tianjin Medical University Cancer Institute, Tianjin, China
| | - Dok Hyun Yoon
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Won-Seog Kim
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Haifaa Abdulhaq
- Department of Medicine, University of California San Francisco, Fresno, CA, USA
| | - William Townsend
- Research Department of Haematology, University College London Hospitals, London, UK
| | - Charles Herbaux
- Clinical Haematology, University Hospital of Montpellier, Montpellier, France
| | - Jan M Zaucha
- Department of Haematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Hung Chang
- Chang Gung Medical Foundation - Linkou Branch, Taoyuan City, Taiwan
| | | | - Chan Yoon Cheah
- Sir Charles Gairdner Hospital, Linear Clinical Research and University of Western Australia, Perth, WA, Australia
| | | | | | | | | | | | | | | | | | - Huang Huang
- F Hoffmann-La Roche, Mississauga, ON, Canada
| | | | - Gareth P Gregory
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Khaliulin M, Valiullina A, Petukhov A, Yuan Y, Spada S, Bulatov E. Breaking the shield of solid tumors: a combined approach for enhanced efficacy of CAR-T cells. Cancer Immunol Immunother 2024; 74:3. [PMID: 39487875 PMCID: PMC11531461 DOI: 10.1007/s00262-024-03817-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: 03/30/2024] [Accepted: 08/22/2024] [Indexed: 11/04/2024]
Abstract
The use of chimeric antigen receptor (CAR)-T cells has enhanced the range of available therapeutic modalities in the context of cancer treatment. CAR-T cells have demonstrated considerable efficacy in the targeted eradication of blood cancer cells, thereby stimulating substantial interest in the advancement of such therapeutic approaches. However, the efficacy of CAR-T cells against solid tumor cells has been limited due to the presence of various obstacles. Solid tumors exhibit antigenic diversity and an immunosuppressive microenvironment, which presents a challenge for immune cells attempting to penetrate the tumor. CAR-T cells also demonstrate decreased proliferative activity and cytotoxicity. Furthermore, concerns exist regarding tumor antigen loss and therapy-associated toxicity. Currently, scientists are working to enhance the structure of the CAR and improve the survival and efficiency of CAR-T cells in recognizing tumor antigens in solid tumors. Chemotherapy drugs are frequently employed in the treatment of malignant neoplasms and can also be used prior to cell therapy to enhance CAR-T cell engraftment. Recent studies have demonstrated that chemotherapy drugs can mitigate the suppressive impact of TME, eliminate the physical barrier by destroying the tumor stroma, and facilitate greater penetration of immune cells and CAR-T cells into the tumor. This, in turn, increases their survival, persistence, and cytotoxicity, as well as affects the metabolism of immune cells inside the tumor. However, the effectiveness of the combined approach against solid tumors depends on several factors, including the type of tumor, dosage, population of CAR-T cells, and individual characteristics of the body. This review examines the principal obstacles to the utilization of CAR-T cells against solid tumors, proposes solutions to these issues, and assesses the potential advantages of a combined approach to radiation exposure, which has the potential to enhance the sensitivity of the tumor to other agents.
Collapse
Affiliation(s)
- Marat Khaliulin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008
| | - Aygul Valiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008
| | - Alexey Petukhov
- Nazarbaev University, Qabanbay Batyr Ave 53, 010000, Astana, Kazakhstan
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, People's Republic of China
| | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia, 117997.
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia, 119048.
| |
Collapse
|
4
|
Arab A, Kashani B, Cordova-Delgado M, Scott EN, Alemi K, Trueman J, Groeneweg G, Chang WC, Loucks CM, Ross CJD, Carleton BC, Ester M. Machine learning model identifies genetic predictors of cisplatin-induced ototoxicity in CERS6 and TLR4. Comput Biol Med 2024; 183:109324. [PMID: 39488053 DOI: 10.1016/j.compbiomed.2024.109324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Cisplatin-induced ototoxicity remains a significant concern in pediatric cancer treatment due to its permanent impact on quality of life. Previously, genetic association analyses have been performed to detect genetic variants associated with this adverse reaction. METHODS In this study, a combination of interpretable neural networks and Generative Adversarial Networks (GANs) was employed to identify genetic markers associated with cisplatin-induced ototoxicity. The applied method, BRI-Net, incorporates biological domain knowledge to define the network structure and employs adversarial training to learn an unbiased representation of the data, which is robust to known confounders. Leveraging genomic data from a cohort of 362 cisplatin-treated pediatric cancer patients recruited by the CPNDS (Canadian Pharmacogenomics Network for Drug Safety), this model revealed two statistically significant single nucleotide polymorphisms to be associated with cisplatin-induced ototoxicity. RESULTS Two markers within the CERS6 (rs13022792, p-value: 3 × 10-4) and TLR4 (rs10759932, p-value: 7 × 10-4) genes were associated with this cisplatin-induced adverse reaction. CERS6, a ceramide synthase, contributes to elevated ceramide levels, a known initiator of apoptotic signals in mouse models of inner ear hair cells. TLR4, a pattern-recognition protein, initiates inflammation in response to cisplatin, and reduced TLR4 expression has been shown in murine hair cells to confer protection from ototoxicity. CONCLUSION Overall, these findings provide a foundation for understanding the genetic landscape of cisplatin-induced ototoxicity, with implications for improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Ali Arab
- School of Computing Science, Simon Fraser University, Burnaby, BC, Canada
| | - Bahareh Kashani
- Department of Experimental Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | | | - Erika N Scott
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kaveh Alemi
- School of Computing Science, Simon Fraser University, Burnaby, BC, Canada
| | - Jessica Trueman
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gabriella Groeneweg
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada
| | - Wan-Chun Chang
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Catrina M Loucks
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Colin J D Ross
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada.
| | - Martin Ester
- School of Computing Science, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
5
|
Peng Z, Huang W, Xiao Z, Wang J, Zhu Y, Zhang F, Lan D, He F. Immunotherapy in the Fight Against Bone Metastases: A Review of Recent Developments and Challenges. Curr Treat Options Oncol 2024; 25:1374-1389. [PMID: 39436492 PMCID: PMC11541271 DOI: 10.1007/s11864-024-01256-7] [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: 08/14/2024] [Indexed: 10/23/2024]
Abstract
OPINION STATEMENT Bone metastasis, a frequent and detrimental complication of advanced cancers, often triggers bone deterioration events that severely compromise patient quality of life and prognosis. The past few years have witnessed the emergence and continuous advancements in immunotherapy, ushering in innovative therapeutic prospects for bone metastasis. These advancements include not only the use of immune checkpoint inhibitors (ICIs), both as standalone and combined treatments, but also the investigation of novel targets within immune cells residing in bone metastases. These breakthroughs have instilled fresh optimism for effectively managing patients with bone metastasis. This article endeavors to present an exhaustive review of the recent progress made across a spectrum of immunotherapeutic strategies and targeted therapies specifically designed for individuals battling bone metastasis from malignant tumors. By doing so, it seeks to offer insights that can inform clinical practices and guide further medical research in this domain.
Collapse
Affiliation(s)
- Zhonghui Peng
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Wei Huang
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Ziyu Xiao
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Jinge Wang
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Yongzhe Zhu
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Fudou Zhang
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Dongqiang Lan
- Department of Oncology, Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, China
| | - Fengjiao He
- Department of Oncology, Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, China.
| |
Collapse
|
6
|
Zhao R, Zhou J, Zheng Z, Xiong X, Wang Q, Li S, Wei W, Guo R. FOLFOX-Based Hepatic Arterial Infusion Chemotherapy with Sequential Drug-Eluting Bead Transarterial Chemoembolization for Unresectable Large Hepatocellular Carcinoma: A Single-Center Retrospective Cohort Study. J Hepatocell Carcinoma 2024; 11:2087-2099. [PMID: 39493266 PMCID: PMC11529345 DOI: 10.2147/jhc.s493577] [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: 09/10/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Background For patients with large unresectable hepatocellular carcinoma (HCC), the effectiveness of conventional transarterial chemoembolization (TACE) remains suboptimal, which necessitates the administration of substantial volumes of chemotherapy drugs and lipiodol, thereby increasing the risk of liver failure and other chemotherapy-related complications. Therefore, we devised a strategy of initial hepatic arterial infusion chemotherapy (HAIC) followed by sequential drug-eluting bead TACE (DEB-TACE). In our treatment design, a lower tumor burden after HAIC facilitated complete embolization of tumor vasculature, and the use of less amount of embolic agents reduced the incidence of liver failure and embolization syndromes. Methods This retrospective study evaluated consecutive patients with unresectable large HCC with a maximum tumor diameter of ≥7 cm who received FOLFOX-HAIC combined with sequential DEB-TACE from April 2019 to February 2024. Efficacy was evaluated using the objective response rate (ORR), overall survival (OS), and progression-free survival (PFS); and safety was assessed using the frequency of key adverse events (AEs). Results Among the 76 patients included, the median maximum tumor diameter was 12.4 cm (range, 7.0-23.4 cm). The overall ORRs based on mRECIST and RECIST 1.1 criteria were 94.1% and 51.5%, respectively. The median OS was 28.1 months (95% CI, 22.7-33.4), and the median PFS was 11.7 months (95% CI, 7.7-15.8). All patients experienced AEs, but only 18.4% experienced grade 3 or 4 AEs, there was no treatment-related mortality. Conclusion In this single-center, retrospective study, our results suggested that FOLFOX-HAIC with sequential DEB-TACE demonstrated promising efficacy and safety for patients with unresectable HCC with a maximum tumor diameter of ≥7 cm.
Collapse
Affiliation(s)
- Rongce Zhao
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Jing Zhou
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Zehao Zheng
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Xinhao Xiong
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Qiaoxuan Wang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Shaohua Li
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Wei Wei
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Rongping Guo
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| |
Collapse
|
7
|
Ogasawara A, Matsushita H, Tan TZ, Shintani D, Ye J, Nagao S, Demachi-Okamura A, Muraoka D, Kobayashi Y, Kakimi K, Yamaguchi R, Matsuo K, Yamamoto K, Fujiwara K, Huang RYJ, Tan DSP, Hasegawa K. Immunological impact of intraperitoneal and intravenous chemotherapy in ovarian cancer, translational analyses of the Phase 3 iPocc trial. Gynecol Oncol 2024; 191:124-131. [PMID: 39413557 DOI: 10.1016/j.ygyno.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The iPocc trial, a randomized, global phase 3 study that compared intraperitoneal (IP) and intravenous (IV) carboplatin with dose-dense paclitaxel chemotherapy in epithelial ovarian cancer (EOC) patients, demonstrated improved progression-free survival in patients who received IP chemotherapy. The present study aimed to investigate the role of preexisting tumor immunity in the clinical outcomes of patients receiving IP chemotherapy. METHODS This study involved analyzing patient data from the iPocc trial, selectively of those whose tumor specimens were preserved at the time of primary surgery. A total of 116 cases ((IP; n = 59), (IV; n = 57)) were subjected to microarray analysis. Single-sample gene set enrichment analyses were performed to evaluate the tumor immune microenvironment. RESULTS Patients with enhanced tumor infiltration of T cells, natural killer (NK) cells, and cytotoxic lymphocytes in the IP group had a longer overall survival (OS) than those in the IV group, but not in the group with low infiltration. IP therapy improved the OS of patients with high expression of immune-related genes such as CD8A and FOXP3. In patients' subdivided into "immune Hot" and "immune Cold" groups based on hierarchical clustering analysis using four parameters representing "Innate immunity," "T cells," "IFNG response" and "Inhibitory molecules," IP therapy significantly improved prognosis in the "immune Hot" group, but not in the "immune Cold" group compared to that of IV therapy. CONCLUSIONS IP chemotherapy enhances the survival rates of patients with EOC with an immune-Hot phenotype in the tumor microenvironment prior to treatment. (Japan Registry of Clinical Trials number, jRCTs031180141.).
Collapse
Affiliation(s)
- Aiko Ogasawara
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan; Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan.
| | - Tuan Zea Tan
- Genomics and Data Analytics Core, Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Daisuke Shintani
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Jieru Ye
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Shoji Nagao
- Gynecologic Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Daisuke Muraoka
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yukari Kobayashi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan; Department of Immunology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei City, Taiwan; Department of Obstetrics & Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Shao Peng Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore; NUS Centre for Cancer Research (N2CR) National University of Singapore, Singapore, Singapore
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan.
| |
Collapse
|
8
|
Kang R, Ma J, Ai B, Liu J, Zheng Z, Yu J. Efficacy and safety of immunotherapy combined with chemotherapy in patients with ES-SCLC: A systematic review and network meta-analysis of RCTs and RWSs. Thorac Cancer 2024. [PMID: 39397360 DOI: 10.1111/1759-7714.15458] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 10/15/2024] Open
Abstract
OBJECTIVES To evaluate the efficacy and safety of programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors in the treatment of extensive-stage small-cell lung cancer (ES-SCLC), we conducted a systematic review and meta-analysis that included randomized controlled trials (RCTs) and real-world studies (RWS). METHODS By scanning PubMed, Web of science, Embase, and other relevant clinical information public databases, nine RCTs and eight RWSs involving 5205 patients were included in the study. We directly compared the differences between chemotherapy and PD-1/PD-L1 inhibitors plus chemotherapy, and determined the optimal treatment strategy through network meta-analysis (NMA). RESULTS Compared to chemotherapy, the addition of PD-1/PD-L1 inhibitors significantly improves the overall survival (OS), progression-free survival (PFS), and objective response rate (ORR) in SCLC patients. Regarding safety, both RCTs and RWSs indicated no significant difference in grade 3-4 adverse events between chemotherapy and chemoimmunotherapy. NMA showed serplulimab plus chemotherapy (Serp_Chemo) appears to provide the best OS, PFS, and ORR benefit, while nivolumab plus chemotherapy shows higher toxicity than other regimens. In subgroup analysis, for elderly patients (age ≥65) and non-elderly (age <65) patients, the most promising quality regimens for achieving better OS extension are atezolizumab plus chemotherapy (Atez_Chemo) and Serp_Chemo, respectively. For patients with PD-L1 ≥ 1% and lactate dehydrogenase (LDH) > upper limit of normal (ULN), there is no apparent OS benefit from immune therapy. CONCLUSIONS In ES-SCLC treatment, adding PD-1/PD-L1 inhibitors to standard chemotherapy improves OS, PFS, and ORR, with Serp_Chemo shows the most promise. Atez_Chemo and Serp_Chemo provided better survival for elderly and non-elderly patients, respectively.
Collapse
Affiliation(s)
- Runting Kang
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, P.R. China
| | - Junling Ma
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, P.R. China
| | - Bin Ai
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, P.R. China
| | - Juanjuan Liu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, P.R. China
| | - Zitong Zheng
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Jiangyong Yu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, P.R. China
| |
Collapse
|
9
|
Wang M, Li F, Wang Z, Lv L, Liu W. Research progress of natural product-conjugated platinum and gold complexes as potential antitumor agents. Eur J Med Chem 2024; 280:116956. [PMID: 39413444 DOI: 10.1016/j.ejmech.2024.116956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Cancer is widely recognized as a serious disease that poses a significant threat to human life and health. The distinctive chemical properties and pronounced antiproliferative activity of platinum drugs are considered to be responsible for their remarkable efficacy in clinical applications. However, undesirable side effects and resistance have severely hampered the treatment of various types of cancer with platinum-based drugs. Natural products (NPs) exhibit extensive pharmacological activities and represent an important source for developing cancer therapeutics. Therefore, the combination of metals and NPs is an attractive strategy for the development of new anticancer agents. Several studies have indicated that combining metals with NPs has a synergistic enhancement effect in antitumor activity. For transition metals, there has been burgeoning research output investigating NP-conjugated platinum and gold complexes. The present article reviews the progress made over the past 5-10 years on the development of NP-conjugated platinum and gold complexes, including a brief introduction to their chemistry and mechanism of action, and a summary of their benefits.
Collapse
Affiliation(s)
- Meiyu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Fuwei Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhaoran Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Lin Lv
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Wukun Liu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
10
|
Porter LH, Harrison SG, Risbridger GP, Lister N, Taylor RA. Left out in the cold: Moving beyond hormonal therapy for the treatment of immunologically cold prostate cancer with CAR T cell immunotherapies. J Steroid Biochem Mol Biol 2024; 243:106571. [PMID: 38909866 DOI: 10.1016/j.jsbmb.2024.106571] [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: 05/13/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
Prostate cancer is primarily hormone-dependent, and medical treatments have focused on inhibiting androgen biosynthesis or signaling through various approaches. Despite significant advances with the introduction of androgen receptor signalling inhibitors (ARSIs), patients continue to progress to castration-resistant prostate cancer (CRPC), highlighting the need for targeted therapies that extend beyond hormonal blockade. Chimeric Antigen Receptor (CAR) T cells and other engineered immune cells represent a new generation of adoptive cellular therapies. While these therapies have significantly enhanced outcomes for patients with hematological malignancies, ongoing research is exploring the broader use of CAR T therapy in solid tumors, including advanced prostate cancer. In general, CAR T cell therapies are less effective against solid cancers with the immunosuppressive tumor microenvironment hindering T cell infiltration, activation and cytotoxicity following antigen recognition. In addition, inherent tumor heterogeneity exists in patients with advanced prostate cancer that may prevent durable therapeutic responses using single-target agents. These barriers must be overcome to inform clinical trial design and improve treatment efficacy. In this review, we discuss the innovative and rationally designed strategies under investigation to improve the clinical translation of cellular immunotherapy in prostate cancer and maximise therapeutic outcomes for these patients.
Collapse
Affiliation(s)
- L H Porter
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - S G Harrison
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - G P Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cabrini Institute, Cabrini Health, Malvern, VIC 3144, Australia
| | - Natalie Lister
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - R A Taylor
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cabrini Institute, Cabrini Health, Malvern, VIC 3144, Australia; Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
11
|
Jiang T, Chen J, Wang H, Wu F, Chen X, Su C, Zhang H, Zhou F, Yang Y, Zhang J, Sun H, Zhang H, Zhou C, Ren S. Genomic correlates of the response to first-line PD-1 blockade plus chemotherapy in patients with advanced non-small-cell lung cancer. Chin Med J (Engl) 2024:00029330-990000000-01186. [PMID: 39164816 PMCID: PMC11407809 DOI: 10.1097/cm9.0000000000003094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Programmed death 1 (PD-1) blockade plus chemotherapy has become the new first-line standard of care for patients with advanced non-small-cell lung cancer (NSCLC). Yet not all NSCLC patients benefit from this regimen. This study aimed to investigate the predictors of PD-1 blockade plus chemotherapy in untreated advanced NSCLC. METHODS We integrated clinical, genomic, and survival data from 287 patients with untreated advanced NSCLC who were enrolled in one of five registered phase 3 trials and received PD-1 blockade plus chemotherapy or chemotherapy alone. We randomly assigned these patients into a discovery cohort (n = 125), a validation cohort (n = 82), and a control cohort (n = 80). The candidate genes that could predict the response to PD-1 blockade plus chemotherapy were identified using data from the discovery cohort and their predictive values were then evaluated in the three cohorts. Immune deconvolution was conducted using transcriptome data of 1014 NSCLC patients from The Cancer Genome Atlas dataset. RESULTS A genomic variation signature, in which one or more of the 15 candidate genes were altered, was correlated with significantly inferior response rates and survival outcomes in patients treated with first-line PD-1 blockade plus chemotherapy in both discovery and validation cohorts. Its predictive value held in multivariate analyses when adjusted for baseline parameters, programmed cell death ligand 1 (PD-L1) expression level, and tumor mutation burden. Moreover, applying both the 15-gene panel and PD-L1 expression level produced better performance than either alone in predicting benefit from this treatment combination. Immune landscape analyses revealed that tumors with one or more variation in the 15-gene panel were associated with few immune infiltrates, indicating an immune-desert tumor microenvironment. CONCLUSION These findings indicate that a 15-gene panel can serve as a negative prediction biomarker for first-line PD-1 blockade plus chemotherapy in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Jian Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Haiping Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Ying Yang
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
| | - Jiao Zhang
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
| | - Huaibo Sun
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
| | - Henghui Zhang
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; School of Oncology, Capital Medical University, Beijing 100038, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
12
|
How JA, Dang M, Lee S, Fellman B, Westin SN, Sood AK, Fleming ND, Shafer A, Yuan Y, Liu J, Zhao L, Celestino J, Hajek R, Morgan MB, Parra ER, Laberiano Fernandez CD, Arrechedera CA, Solis Soto LM, Schmeler KM, Nick A, Lu KH, Coleman R, Wang L, Jazaeri AA. Pembrolizumab plus chemotherapy in frontline treatment of advanced ovarian cancer: Clinical and translational results from a phase 2 trial. MED 2024:S2666-6340(24)00303-9. [PMID: 39151421 DOI: 10.1016/j.medj.2024.07.022] [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: 03/26/2024] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND The efficacy and feasibility of pembrolizumab combined with chemotherapy in frontline management of advanced high-grade epithelial ovarian cancer (EOC) is unknown. Additionally, modification of the tumor microenvironment following neoadjuvant therapy is not well understood. METHODS In this single-arm phase 2 trial (this study was registered at ClinicalTrials.gov: NCT02520154), eligible patients received up to 4 cycles of neoadjuvant chemotherapy followed by interval cytoreduction, 3 cycles of adjuvant intravenous carboplatin/weekly paclitaxel/pembrolizumab, and finally maintenance pembrolizumab until progression or toxicity (maximum 20 cycles). The primary endpoint was progression-free survival (PFS). Secondary endpoints included feasibility, toxicity, and overall survival (OS). PD-L1 staining, multiplex immunofluorescence staining, RNA sequencing, reverse-phase protein array analyses were performed on pre- and post-chemotherapy samples. FINDINGS Thirty-one eligible patients were enrolled. Median PFS and OS was 14.88 (95% CI 12.39-23.00) and 57.43 months (95% CI 30.88-not reached), respectively. Among those with PD-L1 combined positive score (CPS) ≥10, the median PFS and OS were not reached compared to those with CPS <10 (10.50 and 30.90 months, respectively). Feasibility was met, with all patients completing their planned adjuvant cycles. Treatment discontinuation due to immune-related toxicity occurred in 6 patients (20%). Chemotherapy resulted in an infiltration of anti-tumor immune cells in the tumor microenvironment. Samples of patients with the best PFS demonstrated increased expression of NF-κB, TGF-β, and β-catenin signaling. CONCLUSIONS Pembrolizumab with chemotherapy was feasible and resulted in PFS within the historical range for this EOC population. Patients with CPS ≥10 may benefit more from this regimen, and future studies should investigate this potential biomarker. FUNDING This investigator-initiated trial was funded by Merck.
Collapse
Affiliation(s)
- Jeffrey A How
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Minghao Dang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanghoon Lee
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole D Fleming
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aaron Shafer
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Hajek
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margaret B Morgan
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caddie D Laberiano Fernandez
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Claudio A Arrechedera
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa Maren Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kathleen M Schmeler
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Okano S. Immunotherapy for head and neck cancer: Fundamentals and therapeutic development. Auris Nasus Larynx 2024; 51:684-695. [PMID: 38729034 DOI: 10.1016/j.anl.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: 12/26/2023] [Revised: 04/03/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024]
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) has been treated by multidisciplinary therapy consisting of surgery, radiotherapy, and cancer chemotherapy, but the recent advent of immunotherapy has produced significant changes in treatment systems and the results of these therapies. Immunotherapy has greatly improved the outcome of recurrent metastatic SCCHN, and the development of new treatment methods based on immunotherapy is now being applied not only to recurrent metastatic cases but also to locally advanced cases. To understand and practice cancer immunotherapy, it is important to understand the immune environment surrounding cancer, and the changes to which it is subject. Currently, the anti-PD-1 antibody drugs nivolumab and pembrolizumab are the only immunotherapies with proven efficacy in head and neck cancer. However, anti-PD-L1 and anti-CTLA-4 antibody drugs have also been shown to be useful in other types of cancer and are being incorporated into clinical practice. In head and neck cancer, numerous clinical trials have aimed to improve efficacy and safety by combining immunotherapy with other drug therapies and treatment modalities. Combinations of immunotherapy with cancer drugs with different mechanisms of action (cytotoxic agents, molecular-targeted agents, immune checkpoint inhibitors), as well as with radiation therapy and surgery are being investigated, and have the potential to significantly change medical care for these patients. The application of cancer immunotherapy not only to daily clinical practice but also to further therapeutic development requires a clear and complete understanding of the fundamentals of cancer immunotherapy, and knowledge of the numerous clinical studies conducted, both past and present. The results of these trials are numerous, both positive and negative, and a comprehensive understanding of this wide range of completed and ongoing clinical trials is critical to a systematic and comprehensive understanding of their scope and lessons learnt. In this article, after outlining the concepts of ``cancer immune cycle,'' ``cancer immune editing,'' and ``tumor microenvironment'' to provide an understanding of the basics of cancer immunity, we summarize the basics and clinical trial data on representative immune checkpoint inhibitors used in various cancer types, as well as recent therapeutic developments in cancer immunotherapy and the current status of these new treatments.
Collapse
Affiliation(s)
- Susumu Okano
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan.
| |
Collapse
|
14
|
Birnboim-Perach R, Benhar I. Using Combination therapy to overcome diverse challenges of Immune Checkpoint Inhibitors treatment. Int J Biol Sci 2024; 20:3911-3922. [PMID: 39113705 PMCID: PMC11302893 DOI: 10.7150/ijbs.93697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 08/10/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have heralded a new era in immunotherapy, representing a pivotal breakthrough in cancer treatment. Their impact is profound, with ICIs standing as some of the most prescribed anticancer therapies today. Notably, their ability to induce long-term remission even after treatment cessation provides genuine hope for achieving durable cures. However, despite these strides, challenges persist in the landscape of oncology, including resistance phenomena, immune-related adverse events, and suboptimal response rates. In response to these challenges, combination therapy emerges as a promising approach, poised to enhance treatment outcomes and address limitations inherent to single-agent ICI therapy. By synergistically targeting multiple pathways, combination therapy holds the potential to augment therapeutic efficacy while mitigating toxicity and impeding the emergence of resistance mechanisms. Understanding the intricacies underlying resistance development and adverse events is paramount in devising novel and refined combination strategies. A timeline showing FDA approvals of ICIs combination is shown in Figure 1. This review aims to provide a comprehensive and up-to-date examples of different combined therapy strategies that can be used to overcome various challenges regarding ICI treatment. Through the exploration of innovative therapeutic combinations, we aim to provide clinicians and researchers with actionable knowledge to optimize patient outcomes and propel the field of immuno-oncology forward.
Collapse
Affiliation(s)
- Racheli Birnboim-Perach
- Department of Molecular Microbiology and Biotechnology, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Itai Benhar
- Department of Molecular Microbiology and Biotechnology, The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel-Aviv 69978, Israel
- The Tel Aviv University Center for Combatting Pandemics, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
15
|
Li Y, Mondaza-Hernandez JL, Moura DS, Revenko AS, Tolentino A, Nguyen JT, Tran N, Meyer CA, Merino-Garcia J, Ramos R, Di Lernia D, Martin-Broto J, Hayenga HN, Bleris L. STAT6-targeting antisense oligonucleotides against solitary fibrous tumor. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102154. [PMID: 38511173 PMCID: PMC10950871 DOI: 10.1016/j.omtn.2024.102154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024]
Abstract
Solitary fibrous tumor (SFT) is a rare, non-hereditary soft tissue sarcoma thought to originate from fibroblastic mesenchymal stem cells. The etiology of SFT is thought to be due to an environmental intrachromosomal gene fusion between NGFI-A-binding protein 2 (NAB2) and signal transducer and activator protein 6 (STAT6) genes on chromosome 12, wherein the activation domain of STAT6 is fused with the DNA-binding domain of NAB2 resulting in the oncogenesis of SFT. All NAB2-STAT6 fusion variations discovered in SFTs contain the C-terminal of STAT6 transcript, and thus can serve as target site for antisense oligonucleotides (ASOs)-based therapies. Indeed, our in vitro studies show the STAT6 3' untranslated region (UTR)-targeting ASO (ASO 993523) was able to reduce expression of NAB2-STAT6 fusion transcripts in multiple SFT cell models with high efficiency (half-maximal inhibitory concentration: 116-300 nM). Encouragingly, in vivo treatment of SFT patient-derived xenograft mouse models with ASO 993523 resulted in acceptable tolerability profiles, reduced expression of NAB2-STAT6 fusion transcripts in xenograft tissues (21.9%), and, importantly, reduced tumor growth (32.4% decrease in tumor volume compared with the untreated control). Taken together, our study established ASO 993523 as a potential agent for the treatment of SFTs.
Collapse
Affiliation(s)
- Yi Li
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jose L. Mondaza-Hernandez
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), 28049 Madrid, Spain
- University Hospital General de Villalba, 28400 Madrid, Spain
| | - David S. Moura
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), 28049 Madrid, Spain
| | - Alexey S. Revenko
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA 92010, USA
| | - Angelica Tolentino
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA 92010, USA
| | - John T. Nguyen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Nam Tran
- Neurosurgical Oncology, Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Clark A. Meyer
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jose Merino-Garcia
- Pathology Department, University Hospital Fundacion Jimenez Diaz, Universidad Autonoma, Av. Reyes Catolicos 2, 28040 Madrid, Spain
| | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, 07120 Palma de Mallorca, Spain
| | - Davide Di Lernia
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), 28049 Madrid, Spain
| | - Javier Martin-Broto
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), 28049 Madrid, Spain
- University Hospital General de Villalba, 28400 Madrid, Spain
- Medical Oncology Department, University Hospital Fundacion Jimenez Diaz, 28040 Madrid, Spain
| | - Heather N. Hayenga
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Leonidas Bleris
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
16
|
Sun YT, Lu SX, Lai MY, Yang X, Guan WL, Yang LQ, Li YH, Wang FH, Yang DJ, Qiu MZ. Clinical outcomes and biomarker exploration of first-line PD-1 inhibitors plus chemotherapy in patients with low PD-L1-expressing of gastric or gastroesophageal junction adenocarcinoma. Cancer Immunol Immunother 2024; 73:144. [PMID: 38832979 PMCID: PMC11150231 DOI: 10.1007/s00262-024-03721-6] [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: 03/18/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND The beneficial effects of first-line programmed death-1 (PD-1) inhibitors plus chemotherapy in patients with low programmed death-ligand 1 (PD-L1)-expressing advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma are controversial. METHODS We conducted a retrospective analysis of patients with G/GEJ adenocarcinoma who had undergone first-line treatment with PD-1 inhibitors plus chemotherapy between October 2017 and May 2022. The primary outcomes were objective response rate (ORR) and progression-free survival (PFS). SPSS software V27.0 was used for data analysis. RESULTS Of 345 enrolled patients, 290 had measurable lesions. The overall ORR was 59.3%. PD-L1 status was available in 171 patients, and 67.8% of them were considered as low PD-L1 expression level (combined positive score (CPS) < 5). Patients with PD-L1 CPS < 5 showed a lower response rate (51.1% vs 70.8%, P = 0.024) and a worse PFS (P = 0.009) compared to those with PD-L1 CPS ≥ 5. In the PD-L1 low-expression cohort, patients with non-diffuse type, GEJ cancer, synchronous metastasis, distant lymph node metastasis, liver metastasis, non-peritoneal metastasis, and HER2 positive were significantly associated with higher response rates to PD-1 inhibitors plus chemotherapy (P < 0.05). The presence of peritoneal metastasis (P = 0.028) and diffuse type (P = 0.046) were identified as independent predictors of poor PFS in multivariate analysis of the PD-L1 CPS < 5 subgroup. When evaluated for correlation with overall survival (OS) in the PD-L1 low-expression subgroup, peritoneal metastasis was found to be the only independent prognostic factor of an increased risk of death (hazard ratio: 2.31, 95% CI 1.09-4.90; P = 0.029). CONCLUSIONS PD-L1 CPS ≥ 5 is significantly associated with improved response and extended PFS in G/GEJ cancer patients treated with a combination of PD-1 inhibitors and chemotherapy. Specific subgroups within the low PD-L1-expressing population, such as those with non-diffuse-type tumors and without peritoneal metastases, may also benefit from immunotherapy combined with chemotherapy.
Collapse
Affiliation(s)
- Yu-Ting Sun
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Shi-Xun Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Ming-Yu Lai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Xia Yang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Wen-Long Guan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Li-Qiong Yang
- Department of Basic Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Yu-Hong Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China
| | - Da-Jun Yang
- Department of Basic Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China.
| | - Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
17
|
Duan M, Leng S, Mao P. Cisplatin in the era of PARP inhibitors and immunotherapy. Pharmacol Ther 2024; 258:108642. [PMID: 38614254 DOI: 10.1016/j.pharmthera.2024.108642] [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: 01/02/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Platinum compounds such as cisplatin, carboplatin and oxaliplatin are widely used in chemotherapy. Cisplatin induces cytotoxic DNA damage that blocks DNA replication and gene transcription, leading to arrest of cell proliferation. Although platinum therapy alone is effective against many tumors, cancer cells can adapt to the treatment and gain resistance. The mechanisms for cisplatin resistance are complex, including low DNA damage formation, high DNA repair capacity, changes in apoptosis signaling pathways, rewired cell metabolisms, and others. Drug resistance compromises the clinical efficacy and calls for new strategies by combining cisplatin with other therapies. Exciting progress in cancer treatment, particularly development of poly (ADP-ribose) polymerase (PARP) inhibitors and immune checkpoint inhibitors, opened a new chapter to combine cisplatin with these new cancer therapies. In this Review, we discuss how platinum synergizes with PARP inhibitors and immunotherapy to bring new hope to cancer patients.
Collapse
Affiliation(s)
- Mingrui Duan
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Shuguang Leng
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| | - Peng Mao
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
18
|
Yu X, You Z, Liu Y, Fang J, Zhao Q, Sun Z, Song Y, Liu J, Sun C. Sex-based immune microenvironmental feature heterogeneity in response to PD-1 blockade in combination with chemotherapy for patients with untreated advanced non-small-cell lung cancer. Cancer Med 2024; 13:e7423. [PMID: 38899854 PMCID: PMC11188036 DOI: 10.1002/cam4.7423] [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: 02/21/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND To investigate the sex-based heterogeneity of immune microenvironmental feature and its impact on the response to first-line PD-1 blockade plus chemotherapy in patients with driver-negative advanced or metastatic non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS A total of 439 patients with advanced NSCLC treated with first-line PD-1 blockade plus chemotherapy or chemotherapy were identified. Differences in clinical outcomes between female and male patients were determined using Kaplan-Meier curves. Neoantigen burden and five immune microenvironmental markers expression including PD-L1, CD4, CD8, FOXP3, and CD68 were compared between two groups. RESULTS Of 175 eligible patients, 89 received PD-1 blockade plus chemotherapy and 86 received first-line chemotherapy. Forty five were women (25.7%) and 130 were men (74.3%). Female patients received first-line PD-1 blockade in combination with chemotherapy had dramatically better ORR (85.2% vs. 53.2%; p = 0.009), PFS (23.7 vs. 7.3 months; p = 0.013), and OS (46.2 vs. 20.0 months; p = 0.004) than males. Treatment outcomes were similar between females and males in chemotherapy group. Multivariate analyses showed that sex was the independent prognostic factor for patients received PD-1 blockade combined with chemotherapy. Although female patients had significantly lower tumor mutational and neoantigen burden than males, pretreatment tumor tissues of female patients had markedly higher CD4, CD4/FOXP3, and CD4/FOXP3/PD-L1 expression level than male patients. CONCLUSIONS Female patients with untreated advanced or metastatic NSCLC would derive a larger benefit from PD-1 blockade in combination with chemotherapy than males. The biological significances of heterogeneity of tumor immune microenvironmental features between them need further investigation.
Collapse
Affiliation(s)
- Xiaofeng Yu
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Zhaolei You
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Ying Liu
- Department of Medical OncologyYantai Yuhuangding HospitalYantaiChina
| | - Jian Fang
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Qi Zhao
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| | - Zhihong Sun
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| | - Yingjian Song
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Jie Liu
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| | - Chengming Sun
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| |
Collapse
|
19
|
Zhao R, Zhou J, Miao Z, Xiong X, Wei W, Li S, Guo R. Efficacy and safety of lenvatinib plus durvalumab combined with hepatic arterial infusion chemotherapy for unresectable intrahepatic cholangiocarcinoma. Front Immunol 2024; 15:1397827. [PMID: 38799453 PMCID: PMC11116590 DOI: 10.3389/fimmu.2024.1397827] [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: 03/08/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Background The prognosis for unresectable intrahepatic cholangiocarcinoma (ICC) is poor and the efficacy of traditional chemotherapy remains unsatisfactory. Hepatic arterial infusion chemotherapy (HAIC) with oxaliplatin, leucovorin, and 5-fluorouracil (FOLFOX) is effective in patients with unresectable ICC. In this study, we determined the preliminary clinical efficacy and safety of lenvatinib plus durvalumab combined with FOLFOX-HAIC in patients with untreated, unresectable ICC. Materials and methods Between July 2021 and July 2023, patients with unresectable ICC who initially received lenvatinib plus durvalumab combined with FOLFOX-HAIC at the Sun Yat-Sen University Cancer Center (SYSUCC) were reviewed for eligibility. Efficacy was evaluated by tumor response rate and survival, and safety was assessed by the frequency of key adverse events (AEs). Results A total of 28 eligible patients were enrolled. The objective response rates (ORRs) based on mRECIST and RECIST 1.1 criteria were 65.2% and 39.1%, respectively. The median OS was 17.9 months (95% CI, 5.7-30.1) and the median PFS was 11.9 months (95% CI, 6.7-17.1). Most patients (92.9%) experienced adverse events (AEs), whereas 46.5% (13/28) experienced grade 3 or 4 AEs. Conclusion Lenvatinib plus durvalumab combined with FOLFOX-HAIC showed promising antitumor activity and manageable AEs in patients with treatment-naive unresectable ICC. This regimen may be suitable as a novel first-line treatment option for this patient population.
Collapse
Affiliation(s)
- Rongce Zhao
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Zhou
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhaoxia Miao
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xinhao Xiong
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Wei
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shaohua Li
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rongping Guo
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
20
|
Zhang Y, Zhang H, Xu H, Wang Y, Feng L, Yi F. Efficacy and safety of hepatic arterial infusion chemotherapy combined with lenvatinib and PD-1 inhibitors for advanced hepatocellular carcinoma with macrovascular invasion. World J Surg Oncol 2024; 22:122. [PMID: 38711095 PMCID: PMC11071192 DOI: 10.1186/s12957-024-03396-4] [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: 11/08/2023] [Accepted: 04/28/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND AND AIMS The prognosis of hepatocellular carcinoma (HCC) with macrovascular invasion(MaVI)is poor, and the treatment is limited. This study aims to explore the efficacy and safety of hepatic arterial infusion chemotherapy (HAIC), combined with lenvatinib and programmed cell death-1(PD-1) inhibitor in the first-line treatment of HCC with MaVI. METHODS From July 2020 to February 2022, we retrospectively analyzed consecutive patients with HCC with MaVI who received hepatic arterial infusion FOLFOX(oxaliplatin, 5-fluorouracil, and leucovorin)combined with lenvatinib and PD-1 inhibitor. The efficacy was evaluated by RECIST 1.1. Kaplan-Meier was used to explore the overall survival and progression-free survival (PFS), and the COX regression model was used to analyze the risk factors of PFS. Adverse events (AEs) were evaluated according to CTCAE5.0. RESULTS Thirty-two patients with HCC complicated with MaVI were recruited from the Second Affiliated Hospital of Nanchang University. Among the patients treated with HAIC combined with lenvatinib and PD-1 inhibitor, ten patients (31.25%) got partial response, eighteen patients (56.25%) maintained stable disease and four patients (12.50%) suffered progressive disease during follow-up; and objective response rate was 31.25%, and disease control rate was 87.5%. The median PFS was 179 days. Univariate and multivariate Cox analysis showed that the extrahepatic metastases and Child-Pugh score were independent prognostic factors of PFS. Twenty-two (68.75%) patients suffered adverse reactions. The main AEs were elevated transaminase (46.87%), thrombocytopenia (40.63%), hypoalbuminemia (28.13%), nausea and vomiting (21.88%), leukopenia (18.76%), abdominal pain (15.63%), hypertension (15.63%) and fever (15.63%). There were seven cases (21.88%) that had grade 3 or above AEs; Among them, two cases with elevated transaminase (6.25%), leukopenia, thrombocytopenia, nausea and vomiting, abdominal pain, and diarrhea occurred in one case respectively. Moreover, no treatment-related death was observed. CONCLUSIONS Hepatic arterial infusion of FOLFOX combined with lenvatinib and PD-1 inhibitor as the first-line treatment for HCC complicated with MaVI is effective, and adverse reactions are tolerable.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, P.R. of China
- Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang, 330006, P.R. of China
| | - Haiyan Zhang
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, P.R. of China
- Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang, 330006, P.R. of China
| | - Haoqian Xu
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, P.R. of China
- Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang, 330006, P.R. of China
| | - Ying Wang
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, P.R. of China
- Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang, 330006, P.R. of China
| | - Long Feng
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, P.R. of China.
- Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang, 330006, P.R. of China.
| | - Fengming Yi
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, P.R. of China.
- Jiangxi Key Laboratory of Clinical and Translational Cancer Research, Nanchang, 330006, P.R. of China.
| |
Collapse
|
21
|
Ma Z, Xiao Z, Yin P, Wen K, Wang W, Yan Y, Lin Z, Li Z, Wang H, Zhang J, Mao K. Comparison of survival benefit and safety between surgery following conversion therapy versus surgery alone in patients with surgically resectable hepatocellular carcinoma at CNLC IIb/IIIa stage: a propensity score matching study. Int J Surg 2024; 110:2910-2921. [PMID: 38353702 DOI: 10.1097/js9.0000000000001193] [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: 11/25/2023] [Accepted: 01/31/2024] [Indexed: 05/16/2024]
Abstract
OBJECTIVE The objective of this study is to evaluate and compare the survival benefit and safety of surgery following conversion therapy versus surgery alone in patients diagnosed with surgically resectable hepatocellular carcinoma (HCC) at China Liver Cancer Staging (CNLC) IIb/IIIa stage. METHODS A total of 95 patients diagnosed with surgically resectable CNLC IIb/IIIa HCC were retrospectively enrolled in our study from November 2018 to December 2022. Among them, 30 patients underwent conversion therapy followed by hepatectomy, while the remaining 65 received surgery alone. The primary endpoint was recurrence-free survival (RFS). Propensity score matching was employed to minimize bias in the retrospective analysis. RESULTS Compared to the surgery alone group, the conversion therapy group demonstrated a significantly prolonged median RFS (17.1 vs. 7.0 months; P =0.014), a reduced incidence of microvascular invasion (MVI, 23.3 vs. 81.5%; P <0.001), and a comparable rate of achieving Textbook Outcome in Liver Surgery (TOLS, 83.3 vs. 76.9%; P =0.476). Multivariate analysis indicated that conversion therapy was independently associated with improved RFS after hepatectomy (HR=0.511, P =0.027). The same conclusions were obtained after propensity score matching. CONCLUSIONS The findings of our study offer preliminary evidence that preoperative conversion therapy significantly prolongs RFS in patients with surgically resectable HCC at CNLC IIb/IIIa stage. Furthermore, combining conversion therapy and hepatectomy represents a relatively safe treatment strategy.
Collapse
Affiliation(s)
- Zifeng Ma
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Zhiyu Xiao
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Pengfei Yin
- Department of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing
| | - Kai Wen
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Weidong Wang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Yongcong Yan
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Zijian Lin
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Zonglin Li
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Haikuo Wang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Jianlong Zhang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| | - Kai Mao
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou
| |
Collapse
|
22
|
Liu Y, Ping L, Song Y, Tang Y, Zheng W, Liu W, Ying Z, Zhang C, Wu M, Feng F, Lin N, Tu M, Zhu J, Xie Y. Camrelizumab plus gemcitabine and oxaliplatin for relapsed or refractory classical Hodgkin lymphoma: a phase II trial. BMC Med 2024; 22:107. [PMID: 38454451 PMCID: PMC10921783 DOI: 10.1186/s12916-024-03329-8] [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: 10/10/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Classical Hodgkin lymphoma (cHL) is a highly curable disease, while novel therapy is needed for refractory or relapsed (R/R) patients. This phase II trial aimed to evaluate the role of camrelizumab plus gemcitabine and oxaliplatin (GEMOX) in R/R cHL patients. METHODS Transplant-eligible patients with R/R cHL were enrolled and received two 14-day cycles of camrelizumab 200 mg intravenously (IV) and two 28-day cycles of camrelizumab 200 mg IV, gemcitabine 1000 mg/m2 IV, and oxaliplatin 100 mg/m2 IV on days 1 and 15. Patients with partial response (PR) or stable disease received an additional cycle of combination therapy. Those who achieved complete response (CR) or PR proceeded to autologous stem cell transplantation (ASCT). The primary endpoint was the CR rate at the end of protocol therapy before ASCT. RESULTS Forty-two patients were enrolled. At the end of protocol therapy, the objective response rate and CR rate were 94.9% (37/39) and 69.2% (27/39) in the evaluable set, and 88.1% (37/42) and 64.3% (27/42) in the full analysis set, respectively. Twenty-nine patients (69.0%) proceeded to ASCT, and 4 of 5 patients with PR achieved CR after ASCT. After a median follow-up of 20.7 months, the 12-month progression-free survival rate was 96.6% and the 12-month overall survival rate was 100%. Grade 3 or higher treatment emergent adverse events occurred in 28.6% of patients (12/42), mainly hematological toxicity. CONCLUSIONS Camrelizumab combined with GEMOX constitutes an effective salvage therapy for R/R cHL, proving to be relatively well-tolerated and facilitating ASCT in most patients, thus promoting sustained remission. TRIAL REGISTRATION ClinicalTrials.gov NCT04239170. Registered on January 1, 2020.
Collapse
Affiliation(s)
- Yanfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Lingyan Ping
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Yongjing Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Wen Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Weiping Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Zhitao Ying
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Chen Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Meng Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Feier Feng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Ningjing Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Meifeng Tu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Jun Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China
| | - Yan Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Fucheng Road 52, Haidian Region, Beijing, 100142, China.
| |
Collapse
|
23
|
Gherman A, Bolundut D, Ecea R, Balacescu L, Curcean S, Dina C, Balacescu O, Cainap C. Molecular Subtypes, microRNAs and Immunotherapy Response in Metastatic Colorectal Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:397. [PMID: 38541123 PMCID: PMC10972200 DOI: 10.3390/medicina60030397] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/10/2024] [Accepted: 02/18/2024] [Indexed: 11/12/2024]
Abstract
Currently, only a limited set of molecular traits are utilized to direct treatment for metastatic CRC (mCRC). The molecular classification of CRC depicts tumor heterogeneity based on gene expression patterns and aids in comprehending the biological characteristics of tumor formation, growth and prognosis. Additionally, it assists physicians in tailoring the therapeutic approach. Microsatellite instability (MSI-H)/deficient mismatch repair proteins (MMRd) status has become a ubiquitous biomarker in solid tumors, caused by mutations or methylation of genes and, in turn, the accumulation of mutations and antigens that subsequently induce an immune response. Immune checkpoint inhibitors (ICI) have recently received approval for the treatment of mCRC with MSI-H/MMRd status. However, certain individuals experience either initial or acquired resistance. The tumor-programmed cell death ligand 1 (PD-L1) has been linked to the ability of CRC to evade the immune system and promote its growth. Through comprehensive research conducted via the PUBMED database, the objectives of this paper were to review the molecular characteristics linked to tumor response in metastatic CRC in light of improved patients' outcomes following ICI therapies as seen in clinical trials and to identify particular microRNAs that can modulate the expression of specific oncoproteins, such as PD-L1, and disrupt the mechanisms that allow the immune system to be evaded.
Collapse
Affiliation(s)
- Alexandra Gherman
- 10th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.G.); (C.C.)
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| | - Dinu Bolundut
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| | - Radu Ecea
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| | - Loredana Balacescu
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
| | - Sebastian Curcean
- 10th Department of Radiation Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
- Department of Radiation Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Constantin Dina
- Department of Anatomy, Faculty of Medicine, Ovidius University, 124 Mamaia Boulevard, 900527 Constanta, Romania
| | - Ovidiu Balacescu
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
| | - Calin Cainap
- 10th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.G.); (C.C.)
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| |
Collapse
|
24
|
Koeneman BJ, Schreibelt G, Gorris MAJ, Hins - de Bree S, Westdorp H, Ottevanger PB, de Vries IJM. Dendritic cell vaccination combined with carboplatin/paclitaxel for metastatic endometrial cancer patients: results of a phase I/II trial. Front Immunol 2024; 15:1368103. [PMID: 38444861 PMCID: PMC10912556 DOI: 10.3389/fimmu.2024.1368103] [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: 01/09/2024] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Background Metastatic endometrial cancer (mEC) continues to have a poor prognosis despite the introduction of several novel therapies including immune checkpoints inhibitors. Dendritic cell (DC) vaccination is known to be a safe immunotherapeutic modality that can induce immunological and clinical responses in patients with solid tumors. Platinum-based chemotherapy is known to act synergistically with immunotherapy by selectively depleting suppressive immune cells. Therefore, we investigated the immunological efficacy of combined chemoimmunotherapy with an autologous DC vaccine and carboplatin/paclitaxel chemotherapy. Study design This is a prospective, exploratory, single-arm phase I/II study (NCT04212377) in 7 patients with mEC. The DC vaccine consisted of blood-derived conventional and plasmacytoid dendritic cells, loaded with known mEC antigens Mucin-1 and Survivin. Chemotherapy consisted of carboplatin/paclitaxel, given weekly for 6 cycles and three-weekly for 3 cycles. The primary endpoint was immunological vaccine efficacy; secondary endpoints were safety and feasibility. Results Production of DC vaccines was successful in five out of seven patients. These five patients started study treatment and all were able to complete the entire treatment schedule. Antigen-specific responses could be demonstrated in two of the five patients who were treated. All patients had at least one adverse event grade 3 or higher. Treatment-related adverse events grade ≥3 were related to chemotherapy rather than DC vaccination; neutropenia was most common. Suppressive myeloid cells were selectively depleted in peripheral blood after chemotherapy. Conclusion DC vaccination can be safely combined with carboplatin/paclitaxel in patients with metastatic endometrial cancer and induces antigen-specific responses in a minority of patients. Longitudinal immunological phenotyping is suggestive of a synergistic effect of the combination.
Collapse
Affiliation(s)
- Bouke J. Koeneman
- Department of Medical BioSciences, Radboudumc, Nijmegen, Netherlands
- Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| | - Gerty Schreibelt
- Department of Medical BioSciences, Radboudumc, Nijmegen, Netherlands
| | - Mark A. J. Gorris
- Department of Medical BioSciences, Radboudumc, Nijmegen, Netherlands
| | | | - Harm Westdorp
- Department of Medical BioSciences, Radboudumc, Nijmegen, Netherlands
- Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| | | | | |
Collapse
|
25
|
Jin S, Guo Y, Wang X. Development of Platinum Complexes for Tumor Chemoimmunotherapy. Chemistry 2024; 30:e202302948. [PMID: 38171804 DOI: 10.1002/chem.202302948] [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: 09/10/2023] [Indexed: 01/05/2024]
Abstract
Platinum complexes are potential antitumor drugs in chemotherapy. Their impact on tumor treatment could be greatly strengthened by combining with immunotherapy. Increasing evidences indicate that the antitumor activity of platinum complexes is not limited to chemical killing effects, but also extends to immunomodulatory actions. This review introduced the general concept of chemoimmunotherapy and summarized the progress of platinum complexes as chemoimmunotherapeutic agents in recent years. Platinum complexes could be developed into inducers of immunogenic cell death, blockers of immune checkpoint, regulators of immune signaling pathway, and modulators of tumor immune microenvironment, etc. The synergy between chemotherapeutic and immunomodulatory effects reinforces the antitumor activity of platinum complexes, and helps them circumvent the drug resistance and systemic toxicity. The exploration of platinum complexes for chemoimmunotherapy may create new opportunities to revive the discovery of metal anticancer drugs.
Collapse
Affiliation(s)
- Suxing Jin
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Yan Guo
- School of Materials and Chemical Engineering, Henan University of Urban Construction, Pingdingshan, 467036, Henan, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
26
|
Zhao M, Guo Z, Zou YH, Li X, Yan ZP, Chen MS, Fan WJ, Li HL, Yang JJ, Chen XM, Xu LF, Zhang YW, Zhu KS, Sun JH, Li JP, Jin Y, Yu HP, Duan F, Xiong B, Yin GW, Lin HL, Ma YL, Wang HM, Gu SZ, Si TG, Wang XD, Zhao C, Yu WC, Guo JH, Zhai J, Huang YH, Wang WY, Lin HF, Gu YK, Chen JZ, Wang JP, Zhang YM, Yi JZ, Lyu N. Arterial chemotherapy for hepatocellular carcinoma in China: consensus recommendations. Hepatol Int 2024; 18:4-31. [PMID: 37864725 DOI: 10.1007/s12072-023-10599-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/17/2023] [Indexed: 10/23/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies and the third leading cause of cancer-related deaths globally. Hepatic arterial infusion chemotherapy (HAIC) treatment is widely accepted as one of the alternative therapeutic modalities for HCC owing to its local control effect and low systemic toxicity. Nevertheless, although accumulating high-quality evidence has displayed the superior survival advantages of HAIC of oxaliplatin, fluorouracil, and leucovorin (HAIC-FOLFOX) compared with standard first-line treatment in different scenarios, the lack of standardization for HAIC procedure and remained controversy limited the proper and safe performance of HAIC treatment in HCC. Therefore, an expert consensus conference was held on March 2023 in Guangzhou, China to review current practices regarding HAIC treatment in patients with HCC and develop widely accepted statements and recommendations. In this article, the latest evidence of HAIC was systematically summarized and the final 22 expert recommendations were proposed, which incorporate the assessment of candidates for HAIC treatment, procedural technique details, therapeutic outcomes, the HAIC-related complications and corresponding treatments, and therapeutic scheme management.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China.
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China.
| | - Zhi Guo
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Ying-Hua Zou
- Department of Interventional and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Ping Yan
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min-Shan Chen
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei-Jun Fan
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hai-Liang Li
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Ji-Jin Yang
- Department of Interventional Radiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiao-Ming Chen
- Department of Interventional Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lin-Feng Xu
- Department of Interventional Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yue-Wei Zhang
- Hepatopancreatbiliary Center, Tsinghua University Affiliated Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Kang-Shun Zhu
- Department of Minimally Invasive Interventional Radiology and Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun-Hui Sun
- Division of Hepatobiliary and Pancreatic Surgery, Hepatobiliary and Pancreatic Interventional Treatment Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Ping Li
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yong Jin
- The Interventional Therapy Department, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai-Peng Yu
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Feng Duan
- Department of Interventional Radiology, The General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Bin Xiong
- Department of Interventional Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guo-Wen Yin
- Department of Interventional Radiology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Lan Lin
- Department of Interventional Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Yi-Long Ma
- Department of Interventional Therapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Hua-Ming Wang
- Department of Interventional Therapy, The Fifth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Shan-Zhi Gu
- Department of Interventional Therapy, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tong-Guo Si
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Xiao-Dong Wang
- Departments of Interventional Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Chang Zhao
- Department of Interventional Therapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Wen-Chang Yu
- Department of Interventional Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jian-Hai Guo
- Departments of Interventional Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Zhai
- Department of Interventional Radiology, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Yong-Hui Huang
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wei-Yu Wang
- Department of Interventional Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hai-Feng Lin
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yang-Kui Gu
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jin-Zhang Chen
- Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Peng Wang
- Department of Oncology, First People's Hospital of Foshan, Foshan Hospital of Sun Yat-Sen University, Foshan, China
| | - Yi-Min Zhang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jun-Zhe Yi
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Ning Lyu
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China
| |
Collapse
|
27
|
Das S. Comparison of Clinical Trial Results of the Recently Approved Immunotherapeutic Drugs for Advanced Biliary Tract Cancers. Rev Recent Clin Trials 2024; 19:81-90. [PMID: 38288802 DOI: 10.2174/0115748871276666240123043710] [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: 10/04/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 06/25/2024]
Abstract
The recently approved immunotherapeutic drugs are Keytruda (pembrolizumab) and Imfinzi (durvalumab) for advanced biliary tract cancers that inhibit PD-1 receptor and PD-L1 ligand, respectively. In this perspective, the results of the two clinical trials, i.e., TOPAZ-1 (NCT03875235) and KEYNOTE-966 (NCT04003636), are critically appraised, compared, and discussed to assess the benefits of these two drugs in the context of the treatment of advanced biliary tract cancers with a focus on PD-L1 status and MIS (microsatellite instability) status and therapy responsiveness in the subgroups. Analyzing the PD-L2 status in biliary tract cancer patients can aid in assessing the prognostic value of PD-L2 expression in determining the clinical response and this may aid in appropriate patient stratification.
Collapse
Affiliation(s)
- Samayita Das
- Department of Public Health, Harvard Medical School, Boston, MA02115, USA
| |
Collapse
|
28
|
He M, Huang Y, Du Z, Lai Z, Ouyang H, Shen J, Wen D, Li Q, Zhang Y, Wei W, Chen M, Xu L, Kan A, Shi M. Lenvatinib, Toripalimab plus FOLFOX Chemotherapy in Hepatocellular Carcinoma Patients with Extrahepatic Metastasis: A Biomolecular Exploratory, Phase II Trial (LTSC). Clin Cancer Res 2023; 29:5104-5115. [PMID: 37819944 DOI: 10.1158/1078-0432.ccr-23-0060] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/05/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE To investigate the efficacy, safety, and biomarkers of systemic chemotherapy with oxaliplatin, leucovorin, and 5-fluorouracil (FOLFOX) in combination with lenvatinib and toripalimab as the first-line treatment for advanced hepatocellular carcinoma (HCC) with extrahepatic metastasis. PATIENTS AND METHODS In this biomolecular exploratory, phase II trial, eligible patients underwent the triple combination therapy of lenvatinib, toripalimab, plus FOLFOX chemotherapy. Primary endpoint was progression-free survival (PFS) rate at 6 months by RECIST v1.1. Single-nucleus RNA sequencing (snRNA-seq) of tumor biopsy samples was performed for exploratory biomarker analyses. RESULTS Between November 19, 2019, and July 4, 2021, 30 patients were enrolled. The primary endpoint was a 6-month PFS rate of 66.7%, with a median PFS of 9.73 months [95% confidence interval (CI), 2.89-16.58]. The median overall survival (OS) was 14.63 months (95% CI, 11.77-17.50), with an objective response rate of 43.3%. Twenty-four (80.0%) patients exhibited high-risk features, among whom the median OS and PFS were 13.7 months (95% CI, 9.24-18.16) and 8.3 months (95% CI, 3.02-13.58), respectively. The most common adverse events were neutropenia, and increased aspartate aminotransferase and alanine aminotransferase levels. Exploratory analyses of snRNA-seq profiles suggested that patients with higher abundance of tumor-infiltrating immune cells were more likely to benefit from this combination. In addition, two subtypes of hepatocytes (AKR1C2+ and CFHR4+ malignant hepatocytes) were associated with reduced clinical benefits. CONCLUSIONS FOLFOX chemotherapy in combination with lenvatinib and toripalimab showed promising antitumor activity with manageable toxicities in advanced HCC with extrahepatic metastasis. AKR1C2+ and CFHR4+ hepatocyte subtypes may be predictive biomarkers of resistance to the combination therapy.
Collapse
Affiliation(s)
- MinKe He
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - YeXing Huang
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - ZeFeng Du
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - ZhiCheng Lai
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hanyue Ouyang
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - JingXian Shen
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - DongSheng Wen
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - QiJiong Li
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - YaoJun Zhang
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wei Wei
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - MinShan Chen
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Li Xu
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Anna Kan
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ming Shi
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
29
|
Huang X, Ren Q, Yang L, Cui D, Ma C, Zheng Y, Wu J. Immunogenic chemotherapy: great potential for improving response rates. Front Oncol 2023; 13:1308681. [PMID: 38125944 PMCID: PMC10732354 DOI: 10.3389/fonc.2023.1308681] [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: 10/06/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
The activation of anti-tumor immunity is critical in treating cancers. Recent studies indicate that several chemotherapy agents can stimulate anti-tumor immunity by inducing immunogenic cell death and durably eradicate tumors. This suggests that immunogenic chemotherapy holds great potential for improving response rates. However, chemotherapy in practice has only had limited success in inducing long-term survival or cure of cancers when used either alone or in combination with immunotherapy. We think that this is because the importance of dose, schedule, and tumor model dependence of chemotherapy-activated anti-tumor immunity is under-appreciated. Here, we review immune modulation function of representative chemotherapy agents and propose a model of immunogenic chemotherapy-induced long-lasting responses that rely on synergetic interaction between killing tumor cells and inducing anti-tumor immunity. We comb through several chemotherapy treatment schedules, and identify the needs for chemotherapy dose and schedule optimization and combination therapy with immunotherapy when chemotherapy dosage or immune responsiveness is too low. We further review tumor cell intrinsic factors that affect the optimal chemotherapy dose and schedule. Lastly, we review the biomarkers indicating responsiveness to chemotherapy and/or immunotherapy treatments. A deep understanding of how chemotherapy activates anti-tumor immunity and how to monitor its responsiveness can lead to the development of more effective chemotherapy or chemo-immunotherapy, thereby improving the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Xiaojun Huang
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qinghuan Ren
- Alberta Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Leixiang Yang
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Di Cui
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenyang Ma
- Department of Internal Medicine of Traditional Chinese Medicine, The Second People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Yueliang Zheng
- Cancer Center, Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Junjie Wu
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
30
|
Jiang T, Jin Q, Wang J, Wu F, Chen J, Chen G, Huang Y, Chen J, Cheng Y, Wang Q, Pan Y, Zhou J, Shi J, Xu X, Lin L, Zhang W, Zhang Y, Liu Y, Fang Y, Feng J, Wang Z, Hu S, Fang J, Shu Y, Cui J, Hu Y, Yao W, Li X, Lin X, Wang R, Wang Y, Shi W, Feng G, Ni J, Mao B, Ren D, Sun H, Zhang H, Chen L, Zhou C, Ren S. HLA-I Evolutionary Divergence Confers Response to PD-1 Blockade plus Chemotherapy in Untreated Advanced Non-Small Cell Lung Cancer. Clin Cancer Res 2023; 29:4830-4843. [PMID: 37449971 DOI: 10.1158/1078-0432.ccr-23-0604] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/13/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE PD-1 blockade plus chemotherapy has become the new standard of care in patients with untreated advanced non-small cell lung cancer (NSCLC), whereas predictive biomarkers remain undetermined. EXPERIMENTAL DESIGN We integrated clinical, genomic, and survival data of 427 NSCLC patients treated with first-line PD-1 blockade plus chemotherapy or chemotherapy from two phase III trials (CameL and CameL-sq) and investigated the predictive and prognostic value of HLA class I evolutionary divergence (HED). RESULTS High HED could predict significantly improved objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) in those who received PD-1 blockade plus chemotherapy [in the CameL trial, ORR: 81.8% vs. 53.2%; P = 0.032; PFS: hazard ratio (HR), 0.47; P = 0.012; OS: HR, 0.40; P = 0.014; in the CameL-sq trial, ORR: 89.2% vs. 62.3%; P = 0.007; PFS: HR, 0.49; P = 0.005; OS: HR, 0.38; P = 0.002], but not chemotherapy. In multivariate analysis adjusted for PD-L1 expression and tumor mutation burden, high HED was independently associated with markedly better ORR, PFS, and OS in both trials. Moreover, the joint utility of HED and PD-L1 expression showed better performance than either alone in predicting treatment benefit from PD-1 blockade plus chemotherapy. Single-cell RNA sequencing of 58,977 cells collected from 11 patients revealed that tumors with high HED had improved antigen presentation and T cell-mediated antitumor immunity, indicating an inflamed tumor microenvironment phenotype. CONCLUSIONS These findings suggest that high HED could portend survival benefit in advanced NSCLC treated with first-line PD-1 blockade plus chemotherapy. See related commentary by Dimou, p. 4706.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Qiqi Jin
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiahao Wang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Jian Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Gongyan Chen
- First Ward of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yunchao Huang
- Department of Thoracic Surgery, Yunnan Cancer Hospital and the Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Kunming, China
| | - Jianhua Chen
- Department of Medical Oncology-Chest (1), Hunan Cancer Hospital, Changsha, China
| | - Ying Cheng
- Department of Medical Oncology, Jilin Cancer Hospital, Changchun, China
| | - QiMing Wang
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Yueyin Pan
- Department of Chemotherapy Oncology, Anhui Provincial Hospital, Hefei, China
| | - Jianying Zhou
- Respiratory Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Jianhua Shi
- Internal Medicine Ward 2, Linyi Cancer Hospital, Linyi, China
| | - Xingxiang Xu
- Respiratory Department, The Northern Jiangsu People's Hospital, Yangzhou, China
| | - LiZhu Lin
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiping Zhang
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China, Medical University, Shenyang, China
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Jifeng Feng
- Department of Thoracic Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhehai Wang
- Department of Respiratory, Shandong Cancer Hospital and Institute, Jinan, China
| | - Sheng Hu
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, China
| | - Jian Fang
- The Second Department of Thoracic Oncology, Beijing Cancer Hospital, Beijing, China
| | - Yongqian Shu
- Department of Oncology, Jiangsu Province Hospital, Nanjing, China
| | - Jiuwei Cui
- Department of Medical Oncology, The First Bethune Hospital of Jilin University, Changchun, China
| | - Yi Hu
- Oncology Department, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Wenxiu Yao
- Department of Thoracic Oncology, Sichuan Provincial Cancer Hospital, Chengdu, China
| | - Xingya Li
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Lin
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui Wang
- Department of Medical Oncology, Anhui Chest Hospital, Hefei, China
| | - Yongsheng Wang
- Department of Thoracic Medical Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Shi
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals, China
| | - Gaohua Feng
- Department of Pulmonary and Critical Care Medicine, Zhangjiagang Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Jun Ni
- Department of Pulmonary and Critical Care Medicine, Zhangjiagang Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Beibei Mao
- Genecast Biotechnology Co., Ltd, Jiangsu Province, China
| | - Dandan Ren
- Genecast Biotechnology Co., Ltd, Jiangsu Province, China
| | - Huaibo Sun
- Genecast Biotechnology Co., Ltd, Jiangsu Province, China
| | - Henghui Zhang
- Genecast Biotechnology Co., Ltd, Jiangsu Province, China
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; School of Oncology, Capital Medical University, Beijing, China
| | - Luonan Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
de Moraes FCA, Pasqualotto E, Lopes LM, Cavalcanti Souza ME, de Oliveira Rodrigues ALS, de Almeida AM, Stecca C, Fernandes MR, Dos Santos NPC. PD-1/PD-L1 inhibitors plus carboplatin and paclitaxel compared with carboplatin and paclitaxel in primary advanced or recurrent endometrial cancer: a systematic review and meta-analysis of randomized clinical trials. BMC Cancer 2023; 23:1166. [PMID: 38031003 PMCID: PMC10688003 DOI: 10.1186/s12885-023-11654-z] [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: 09/06/2023] [Accepted: 11/18/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Paclitaxel and carboplatin is the standard chemotherapy for the treatment of advanced or recurrent endometrial cancer. However, the benefit of adding programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors to chemotherapy is still unclear. METHOD We searched PubMed, Scopus, Cochrane, and Web of Science databases for randomized controlled trials that investigated PD-1/PD-L1 inhibitors plus carboplatin and paclitaxel compared with carboplatin and paclitaxel in primary advanced or recurrent endometrial cancer. We computed hazard ratios (HRs) or risk ratios (RRs) for binary endpoints, with 95% confidence intervals (CIs). We used DerSimonian and Laird random-effect models for all endpoints. Heterogeneity was assessed using I2 statistics. R, version 4.2.3, was used for statistical analyses. RESULTS A total of three studies and 1,431 patients were included. Compared with carboplatin plus paclitaxel-based chemotherapy, progression-free survival (PFS) rate (HR 0.32; 95% CI 0.23-0.44; p < 0.001) and overall survival (OS) at 30 months (RR 3.13; 95% CI 1.26-7.78; p = 0.01) were significant in favor of the PD-1/PD-L1 inhibitors plus carboplatin and paclitaxel group in the mismatch repair-deficient subgroup. However, there were no significant differences in the mismatch repair-proficient subgroup for PFS (HR 0.74; 95% CI 0.50-1.08; p = 0.117) or OS at 30 months (RR 2.24; 95% CI 0.79-6.39; p = 0.13). CONCLUSION Immunotherapy plus carboplatin-paclitaxel increased significantly PFS and OS among patients with advanced or recurrent endometrial cancer, with a significant benefit in the mismatch repair-deficient and high microsatellite instability population.
Collapse
Affiliation(s)
- Francisco Cezar Aquino de Moraes
- Oncology Research Center, Federal University of Pará, University Hospital João de Barros de Barreto, Rua dos Mundurucus, nº4487, Belém, 66073-000, PA, Brazil.
| | - Eric Pasqualotto
- Federal University of Santa Catarina, Florianópolis, 88040-900, Santa Catarina, Brazil
| | | | | | | | | | - Carlos Stecca
- Mackenzie Evangelical University Hospital, Curitiba, 80730-150, Paraná, Brazil
| | - Marianne Rodrigues Fernandes
- Oncology Research Center, Federal University of Pará, University Hospital João de Barros de Barreto, Rua dos Mundurucus, nº4487, Belém, 66073-000, PA, Brazil
| | - Ney Pereira Carneiro Dos Santos
- Oncology Research Center, Federal University of Pará, University Hospital João de Barros de Barreto, Rua dos Mundurucus, nº4487, Belém, 66073-000, PA, Brazil
| |
Collapse
|
32
|
Ding Y, Wang S, Qiu Z, Zhu C, Wang Y, Zhao S, Qiu W, Wang K, Lv J, Qi W. The worthy role of hepatic arterial infusion chemotherapy in combination with anti-programmed cell death protein 1 monoclonal antibody immunotherapy in advanced hepatocellular carcinoma. Front Immunol 2023; 14:1284937. [PMID: 38022559 PMCID: PMC10644007 DOI: 10.3389/fimmu.2023.1284937] [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: 08/29/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Systemic therapy remains the primary therapeutic approach for advanced hepatocellular carcinoma (HCC). Nonetheless, its efficacy in achieving control of intrahepatic lesions is constrained. Hepatic arterial infusion chemotherapy (HAIC) is a therapeutic approach that combines localized treatment with systemic antitumor effects, which aim is to effectively manage the progression of cancerous lesions within the liver, particularly in patients with portal vein tumor thrombosis (PVTT). Combining HAIC with anti-programmed cell death protein 1 (anti-PD-1) monoclonal antibody (mAb) immunotherapy is anticipated to emerge as a novel therapeutic approach aimed at augmenting the response inside the localized tumor site and achieving prolonged survival advantages. In order to assess the effectiveness, safety, and applicability of various therapeutic modalities and to address potential molecular mechanisms underlying the efficacy of HAIC-sensitizing immunotherapy, we reviewed the literature about the combination of HAIC with anti-PD-1 mAb therapies.
Collapse
Affiliation(s)
- Yixin Ding
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Zhenkang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chunyang Zhu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yan Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Shufen Zhao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Kongjia Wang
- Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Weiwei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
33
|
Yao S, Han Y, Yang M, Jin K, Lan H. It's high-time to re-evaluate the value of induced-chemotherapy for reinforcing immunotherapy in colorectal cancer. Front Immunol 2023; 14:1241208. [PMID: 37920463 PMCID: PMC10619163 DOI: 10.3389/fimmu.2023.1241208] [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: 06/16/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
Immunotherapy has made significant advances in the treatment of colorectal cancer (CRC), revolutionizing the therapeutic landscape and highlighting the indispensable role of the tumor immune microenvironment. However, some CRCs have shown poor response to immunotherapy, prompting investigation into the underlying reasons. It has been discovered that certain chemotherapeutic agents possess immune-stimulatory properties, including the induction of immunogenic cell death (ICD), the generation and processing of non-mutated neoantigens (NM-neoAgs), and the B cell follicle-driven T cell response. Based on these findings, the concept of inducing chemotherapy has been introduced, and the combination of inducing chemotherapy and immunotherapy has become a standard treatment option for certain cancers. Clinical trials have confirmed the feasibility and safety of this approach in CRC, offering a promising method for improving the efficacy of immunotherapy. Nevertheless, there are still many challenges and difficulties ahead, and further research is required to optimize its use.
Collapse
Affiliation(s)
- Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yuejun Han
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Mengxiang Yang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Roman V, Mihaila M, Radu N, Marineata S, Diaconu CC, Bostan M. Cell Culture Model Evolution and Its Impact on Improving Therapy Efficiency in Lung Cancer. Cancers (Basel) 2023; 15:4996. [PMID: 37894363 PMCID: PMC10605536 DOI: 10.3390/cancers15204996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Optimizing cell culture conditions is essential to ensure experimental reproducibility. To improve the accuracy of preclinical predictions about the response of tumor cells to different classes of drugs, researchers have used 2D or 3D cell cultures in vitro to mimic the cellular processes occurring in vivo. While 2D cell culture provides valuable information on how therapeutic agents act on tumor cells, it cannot quantify how the tumor microenvironment influences the response to therapy. This review presents the necessary strategies for transitioning from 2D to 3D cell cultures, which have facilitated the rapid evolution of bioengineering techniques, leading to the development of microfluidic technology, including organ-on-chip and tumor-on-chip devices. Additionally, the study aims to highlight the impact of the advent of 3D bioprinting and microfluidic technology and their implications for improving cancer treatment and approaching personalized therapy, especially for lung cancer. Furthermore, implementing microfluidic technology in cancer studies can generate a series of challenges and future perspectives that lead to the discovery of new predictive markers or targets for antitumor treatment.
Collapse
Affiliation(s)
- Viviana Roman
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (V.R.); (M.B.)
| | - Mirela Mihaila
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (V.R.); (M.B.)
| | - Nicoleta Radu
- Department of Biotechnology, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 011464 Bucharest, Romania
- Biotechnology Department, National Institute for Chemistry and Petrochemistry R&D of Bucharest, 060021 Bucharest, Romania
| | - Stefania Marineata
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 050471 Bucharest, Romania;
| | - Carmen Cristina Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania;
| | - Marinela Bostan
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (V.R.); (M.B.)
- Department of Immunology, ‘Victor Babeș’ National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
35
|
Wang RN, Yu Q, Wang XB, Zhu D, Li GL, Li ZX, Jiang W, Li W, Dang YJ. Bis(benzonitrile) dichloroplatinum (II) interrupts PD-1/PD-L1 interaction by binding to PD-1. Acta Pharmacol Sin 2023; 44:2103-2112. [PMID: 37193754 PMCID: PMC10545660 DOI: 10.1038/s41401-023-01092-9] [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: 09/22/2022] [Accepted: 04/10/2023] [Indexed: 05/18/2023] Open
Abstract
Checkpoint inhibitors such as PD-1/PD-L1 antibody therapeutics are a promising option for the treatment of multiple cancers. Due to the inherent limitations of antibodies, great efforts have been devoted to developing small-molecule PD-1/PD-L1 signaling pathway inhibitors. In this study we established a high-throughput AlphaLISA assay to discover small molecules with new skeletons that could block PD-1/PD-L1 interaction. We screened a small-molecule library of 4169 compounds including natural products, FDA approved drugs and other synthetic compounds. Among the 8 potential hits, we found that cisplatin, a first-line chemotherapeutic drug, reduced AlphaLISA signal with an EC50 of 8.3 ± 2.2 μM. Furthermore, we showed that cisplatin-DMSO adduct, but not semplice cisplatin, inhibited PD-1/PD-L1 interaction. Thus, we assessed several commercial platinum (II) compounds, and found that bis(benzonitrile) dichloroplatinum (II) disturbed PD-1/PD-L1 interaction (EC50 = 13.2 ± 3.5 μM). Its inhibitory activity on PD-1/PD-L1 interaction was confirmed in co-immunoprecipitation and PD-1/PD-L1 signaling pathway blockade bioassays. Surface plasmon resonance assay revealed that bis(benzonitrile) dichloroplatinum (II) bound to PD-1 (KD = 2.08 μM) but not PD-L1. In immune-competent wild-type mice but not in immunodeficient nude mice, bis(benzonitrile) dichloroplatinum (II) (7.5 mg/kg, i.p., every 3 days) significantly suppressed the growth of MC38 colorectal cancer xenografts with increasing tumor-infiltrating T cells. These data highlight that platinum compounds are potential immune checkpoint inhibitors for the treatment of cancers.
Collapse
Affiliation(s)
- Rui-Na Wang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Yu
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiao-Bo Wang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Di Zhu
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Guo-Long Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Zeng-Xia Li
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wei Jiang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Wei Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China.
| | - Yong-Jun Dang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
36
|
Yu S, Xiao H, Ma L, Zhang J, Zhang J. Reinforcing the immunogenic cell death to enhance cancer immunotherapy efficacy. Biochim Biophys Acta Rev Cancer 2023; 1878:188946. [PMID: 37385565 DOI: 10.1016/j.bbcan.2023.188946] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Immunogenic cell death (ICD) has been a revolutionary modality in cancer treatment since it kills primary tumors and prevents recurrent malignancy simultaneously. ICD represents a particular form of cancer cell death accompanied by production of damage-associated molecular patterns (DAMPs) that can be recognized by pattern recognition receptors (PRRs), which enhances infiltration of effector T cells and potentiates antitumor immune responses. Various treatment methods can elicit ICD involving chemo- and radio-therapy, phototherapy and nanotechnology to efficiently convert dead cancer cells into vaccines and trigger the antigen-specific immune responses. Nevertheless, the efficacy of ICD-induced therapies is restrained due to low accumulation in the tumor sites and damage of normal tissues. Thus, researchers have been devoted to overcoming these problems with novel materials and strategies. In this review, current knowledge on different ICD modalities, various ICD inducers, development and application of novel ICD-inducing strategies are summarized. Moreover, the prospects and challenges are briefly outlined to provide reference for future design of novel immunotherapy based on ICD effect.
Collapse
Affiliation(s)
- Sihui Yu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Hongyang Xiao
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Li Ma
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Jiarong Zhang
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
37
|
Xu X, Dai F, Mao Y, Zhang K, Qin Y, Zheng J. Metallodrugs in the battle against non-small cell lung cancer: unlocking the potential for improved therapeutic outcomes. Front Pharmacol 2023; 14:1242488. [PMID: 37727388 PMCID: PMC10506097 DOI: 10.3389/fphar.2023.1242488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer mortality worldwide. Platinum-based chemotherapy is standard-of-care but has limitations including toxicity and resistance. Metal complexes of gold, ruthenium, and other metals have emerged as promising alternatives. This review provides a comprehensive analysis of metallodrugs for NSCLC. Bibliometric analysis reveals growing interest in elucidating mechanisms, developing targeted therapies, and synergistic combinations. Classification of metallodrugs highlights platinum, gold, and ruthenium compounds, as well as emerging metals. Diverse mechanisms include DNA damage, redox modulation, and immunomodulation. Preclinical studies demonstrate cytotoxicity and antitumor effects in vitro and in vivo, providing proof-of-concept. Clinical trials indicate platinums have utility but resistance remains problematic. Non-platinum metallodrugs exhibit favorable safety but modest single agent efficacy to date. Drug delivery approaches like nanoparticles show potential to enhance therapeutic index. Future directions include optimization of metal-based complexes, elucidation of resistance mechanisms, biomarker development, and combination therapies to fully realize the promise of metallodrugs for NSCLC.
Collapse
Affiliation(s)
- Xianzhi Xu
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| | - Feng Dai
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yiting Mao
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| | - Kai Zhang
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| | - Ying Qin
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| | - Jiwei Zheng
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
38
|
Colombo I, Karakasis K, Suku S, Oza AM. Chasing Immune Checkpoint Inhibitors in Ovarian Cancer: Novel Combinations and Biomarker Discovery. Cancers (Basel) 2023; 15:3220. [PMID: 37370830 DOI: 10.3390/cancers15123220] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
A deep understanding of the tumor microenvironment and the recognition of tumor-infiltrating lymphocytes as a prognostic factor have resulted in major milestones in immunotherapy that have led to therapeutic advances in treating many cancers. Yet, the translation of this knowledge to clinical success for ovarian cancer remains a challenge. The efficacy of immune checkpoint inhibitors as single agents or combined with chemotherapy has been unsatisfactory, leading to the exploration of alternative combination strategies with targeted agents (e.g., poly-ADP-ribose inhibitors (PARP)and angiogenesis inhibitors) and novel immunotherapy approaches. Among the different histological subtypes, clear cell ovarian cancer has shown a higher sensitivity to immunotherapy. A deeper understanding of the mechanism of immune resistance within the context of ovarian cancer and the identification of predictive biomarkers remain central discovery benchmarks to be realized. This will be critical to successfully define the precision use of immune checkpoint inhibitors for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ilaria Colombo
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Via A. Gallino, 6500 Bellinzona, Switzerland
| | - Katherine Karakasis
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - Sneha Suku
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - Amit M Oza
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
39
|
Li Y, Nguyen JT, Ammanamanchi M, Zhou Z, Harbut EF, Mondaza-Hernandez JL, Meyer CA, Moura DS, Martin-Broto J, Hayenga HN, Bleris L. Reduction of Tumor Growth with RNA-Targeting Treatment of the NAB2-STAT6 Fusion Transcript in Solitary Fibrous Tumor Models. Cancers (Basel) 2023; 15:3127. [PMID: 37370737 DOI: 10.3390/cancers15123127] [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: 05/02/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Solitary fibrous tumor (SFT) is a rare soft-tissue sarcoma. This nonhereditary cancer is the result of an environmental intrachromosomal gene fusion between NAB2 and STAT6 on chromosome 12, which fuses the activation domain of STAT6 with the repression domain of NAB2. Currently there is not an approved chemotherapy regimen for SFTs. The best response on available pharmaceuticals is a partial response or stable disease for several months. The purpose of this study is to investigate the potential of RNA-based therapies for the treatment of SFTs. Specifically, in vitro SFT cell models were engineered to harbor the characteristic NAB2-STAT6 fusion using the CRISPR/SpCas9 system. Cell migration as well as multiple cancer-related signaling pathways were increased in the engineered cells as compared to the fusion-absent parent cells. The SFT cell models were then used for evaluating the targeting efficacies of NAB2-STAT6 fusion-specific antisense oligonucleotides (ASOs) and CRISPR/CasRx systems. Our results showed that fusion specific ASO treatments caused a 58% reduction in expression of fusion transcripts and a 22% reduction in cell proliferation after 72 h in vitro. Similarly, the AAV2-mediated CRISPR/CasRx system led to a 59% reduction in fusion transcript expressions in vitro, and a 55% reduction in xenograft growth after 29 days ex vivo.
Collapse
Affiliation(s)
- Yi Li
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | - John T Nguyen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | | | - Zikun Zhou
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Elijah F Harbut
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jose L Mondaza-Hernandez
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), 28049 Madrid, Spain
- University Hospital General de Villalba, 28400 Madrid, Spain
| | - Clark A Meyer
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - David S Moura
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), 28049 Madrid, Spain
| | - Javier Martin-Broto
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), 28049 Madrid, Spain
- University Hospital General de Villalba, 28400 Madrid, Spain
- Medical Oncology Department, University Hospital Fundación Jimenez Diaz, 28040 Madrid, Spain
| | - Heather N Hayenga
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Leonidas Bleris
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
40
|
Eskander RN, Sill MW, Beffa L, Moore RG, Hope JM, Musa FB, Mannel R, Shahin MS, Cantuaria GH, Girda E, Mathews C, Kavecansky J, Leath CA, Gien LT, Hinchcliff EM, Lele SB, Landrum LM, Backes F, O'Cearbhaill RE, Al Baghdadi T, Hill EK, Thaker PH, John VS, Welch S, Fader AN, Powell MA, Aghajanian C. Pembrolizumab plus Chemotherapy in Advanced Endometrial Cancer. N Engl J Med 2023; 388:2159-2170. [PMID: 36972022 PMCID: PMC10351614 DOI: 10.1056/nejmoa2302312] [Citation(s) in RCA: 218] [Impact Index Per Article: 218.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
BACKGROUND Standard first-line chemotherapy for endometrial cancer is paclitaxel plus carboplatin. The benefit of adding pembrolizumab to chemotherapy remains unclear. METHODS In this double-blind, placebo-controlled, randomized, phase 3 trial, we assigned 816 patients with measurable disease (stage III or IVA) or stage IVB or recurrent endometrial cancer in a 1:1 ratio to receive pembrolizumab or placebo along with combination therapy with paclitaxel plus carboplatin. The administration of pembrolizumab or placebo was planned in 6 cycles every 3 weeks, followed by up to 14 maintenance cycles every 6 weeks. The patients were stratified into two cohorts according to whether they had mismatch repair-deficient (dMMR) or mismatch repair-proficient (pMMR) disease. Previous adjuvant chemotherapy was permitted if the treatment-free interval was at least 12 months. The primary outcome was progression-free survival in the two cohorts. Interim analyses were scheduled to be triggered after the occurrence of at least 84 events of death or progression in the dMMR cohort and at least 196 events in the pMMR cohort. RESULTS In the 12-month analysis, Kaplan-Meier estimates of progression-free survival in the dMMR cohort were 74% in the pembrolizumab group and 38% in the placebo group (hazard ratio for progression or death, 0.30; 95% confidence interval [CI], 0.19 to 0.48; P<0.001), a 70% difference in relative risk. In the pMMR cohort, median progression-free survival was 13.1 months with pembrolizumab and 8.7 months with placebo (hazard ratio, 0.54; 95% CI, 0.41 to 0.71; P<0.001). Adverse events were as expected for pembrolizumab and combination chemotherapy. CONCLUSIONS In patients with advanced or recurrent endometrial cancer, the addition of pembrolizumab to standard chemotherapy resulted in significantly longer progression-free survival than with chemotherapy alone. (Funded by the National Cancer Institute and others; NRG-GY018 ClinicalTrials.gov number, NCT03914612.).
Collapse
Affiliation(s)
- Ramez N Eskander
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Michael W Sill
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Lindsey Beffa
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Richard G Moore
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Joanie M Hope
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Fernanda B Musa
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Robert Mannel
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Mark S Shahin
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Guilherme H Cantuaria
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Eugenia Girda
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Cara Mathews
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Juraj Kavecansky
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Charles A Leath
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Lilian T Gien
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Emily M Hinchcliff
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Shashikant B Lele
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Lisa M Landrum
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Floor Backes
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Roisin E O'Cearbhaill
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Tareq Al Baghdadi
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Emily K Hill
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Premal H Thaker
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Veena S John
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Stephen Welch
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Amanda N Fader
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Matthew A Powell
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| | - Carol Aghajanian
- From the Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, Rebecca and John Moores Cancer Center, La Jolla (R.N.E.), and the Kaiser Permanente National Cancer Institute Community Oncology Research Program (NCORP), Antioch Medical Center, Antioch (J.K.) - both in California; the Clinical Trial Development Division, Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo (M.W.S., S.B.L.), the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester (R.G.M.), the Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York (R.E.O., C.A.), and the Northwell Health Cancer Institute, New Hyde Park (V.S.J.) - all in New York; the Case Comprehensive Cancer Center, Cleveland Clinic Foundation, Cleveland (L.B.), and Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus (F.B.); the Pacific Cancer Research Consortium, NCORP, Alaska Women's Cancer Care, and Providence Alaska Cancer Center, Anchorage (J.M.H.); the Pacific Cancer Research Consortium, NCORP, Swedish Medical Center-First Hill, Seattle (F.B.M.); the University of Oklahoma Health Sciences Center, Oklahoma City (R.M.); Jefferson Abington Hospital, Asplundh Cancer Pavilion of Sidney Kimmel Cancer Center, Jefferson Health, Willow Grove, PA (M.S.S.); Georgia NCORP, Atlanta (G.H.C.); Rutgers Cancer Institute of New Jersey, New Brunswick (E.G.); Women and Infants Hospital, Legoretta Cancer Center, Alpert Medical School, Brown University, Providence, RI (C.M.); the University of Alabama at Birmingham-Deep South Research Consortium, O'Neal Comprehensive Cancer Center, University of Alabama Hospital, Birmingham (C.A.L.); Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto (L.T.G.), and the London Regional Cancer Program, London, ON (S.W.) - both in Canada; Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago (E.M.H.); the Indiana University Health Simon Cancer Center, Indianapolis (L.M.L.); the Michigan Cancer Research Consortium, NCORP, Trinity Health IHA Medical Group, Ypsilanti (T.A.B.); the University of Iowa Hospitals and Clinics, Iowa City (E.K.H.); the Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis (P.H.T., M.A.P.); and the Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore (A.N.F.)
| |
Collapse
|
41
|
Wu C, Spector SA, Theodoropoulos G, Nguyen DJM, Kim EY, Garcia A, Savaraj N, Lim DC, Paul A, Feun LG, Bickerdike M, Wangpaichitr M. Dual inhibition of IDO1/TDO2 enhances anti-tumor immunity in platinum-resistant non-small cell lung cancer. Cancer Metab 2023; 11:7. [PMID: 37226257 DOI: 10.1186/s40170-023-00307-1] [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: 10/17/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND The impact of non-small cell lung cancer (NSCLC) metabolism on the immune microenvironment is not well understood within platinum resistance. We have identified crucial metabolic differences between cisplatin-resistant (CR) and cisplatin-sensitive (CS) NSCLC cells with elevated indoleamine 2,3-dioxygenase-1 (IDO1) activity in CR, recognized by increased kynurenine (KYN) production. METHODS Co-culture, syngeneic, and humanize mice models were utilized. C57BL/6 mice were inoculated with either Lewis lung carcinoma mouse cells (LLC) or their platinum-resistant counterpart (LLC-CR) cells. Humanized mice were inoculated with either A (human CS cells) or ALC (human CR cells). Mice were treated with either IDO1 inhibitor or TDO2 (tryptophan 2,3-dioxygenase-2) inhibitor at 200 mg/kg P.O. once a day for 15 days; or with a new-in-class, IDO1/TDO2 dual inhibitor AT-0174 at 170 mg/kg P.O. once a day for 15 days with and without anti-PD1 antibody (10 mg/kg, every 3 days). Immune profiles and KYN and tryptophan (TRP) production were evaluated. RESULTS CR tumors exhibited a more highly immunosuppressive environment that debilitated robust anti-tumor immune responses. IDO1-mediated KYN production from CR cells suppressed NKG2D on immune effector natural killer (NK) and CD8+ T cells and enhanced immunosuppressive populations of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Importantly, while selective IDO1 inhibition attenuated CR tumor growth, it concomitantly upregulated the TDO2 enzyme. To overcome the compensatory induction of TDO2 activity, we employed the IDO1/TDO2 dual inhibitor, AT-0174. Dual inhibition of IDO1/TDO2 in CR mice suppressed tumor growth to a greater degree than IDO1 inhibition alone. Significant enhancement in NKG2D frequency on NK and CD8+ T cells and a reduction in Tregs and MDSCs were observed following AT-1074 treatment. PD-L1 (programmed death-ligand-1) expression was increased in CR cells; therefore, we assessed dual inhibition + PD1 (programmed cell death protein-1) blocking and report profound anti-tumor growth and improved immunity in CR tumors which in turn extended overall survival in mice. CONCLUSION Our study reports the presence of platinum-resistant lung tumors that utilize both IDO1/TDO2 enzymes for survival, and to escape immune surveillance as a consequence of KYN metabolites. We also report early in vivo data in support of the potential therapeutic efficacy of the dual IDO1/TDO2 inhibitor AT-0174 as a part of immuno-therapeutic treatment that disrupts tumor metabolism and enhances anti-tumor immunity.
Collapse
Affiliation(s)
- Chunjing Wu
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Sydney A Spector
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | | | - Dan J M Nguyen
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Emily Y Kim
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Ashley Garcia
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Niramol Savaraj
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | - Diane C Lim
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | - Ankita Paul
- Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA, USA
| | - Lynn G Feun
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | | | - Medhi Wangpaichitr
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA.
- Department of Surgery, University of Miami School of Medicine, Miami, FL, USA.
| |
Collapse
|
42
|
Alfonso-Triguero P, Lorenzo J, Candiota AP, Arús C, Ruiz-Molina D, Novio F. Platinum-Based Nanoformulations for Glioblastoma Treatment: The Resurgence of Platinum Drugs? NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1619. [PMID: 37242036 PMCID: PMC10223043 DOI: 10.3390/nano13101619] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023]
Abstract
Current therapies for treating Glioblastoma (GB), and brain tumours in general, are inefficient and represent numerous challenges. In addition to surgical resection, chemotherapy and radiotherapy are presently used as standards of care. However, treated patients still face a dismal prognosis with a median survival below 15-18 months. Temozolomide (TMZ) is the main chemotherapeutic agent administered; however, intrinsic or acquired resistance to TMZ contributes to the limited efficacy of this drug. To circumvent the current drawbacks in GB treatment, a large number of classical and non-classical platinum complexes have been prepared and tested for anticancer activity, especially platinum (IV)-based prodrugs. Platinum complexes, used as alkylating agents in the anticancer chemotherapy of some malignancies, are though often associated with severe systemic toxicity (i.e., neurotoxicity), especially after long-term treatments. The objective of the current developments is to produce novel nanoformulations with improved lipophilicity and passive diffusion, promoting intracellular accumulation, while reducing toxicity and optimizing the concomitant treatment of chemo-/radiotherapy. Moreover, the blood-brain barrier (BBB) prevents the access of the drugs to the brain and accumulation in tumour cells, so it represents a key challenge for GB management. The development of novel nanomedicines with the ability to (i) encapsulate Pt-based drugs and pro-drugs, (ii) cross the BBB, and (iii) specifically target cancer cells represents a promising approach to increase the therapeutic effect of the anticancer drugs and reduce undesired side effects. In this review, a critical discussion is presented concerning different families of nanoparticles able to encapsulate platinum anticancer drugs and their application for GB treatment, emphasizing their potential for increasing the effectiveness of platinum-based drugs.
Collapse
Affiliation(s)
- Paula Alfonso-Triguero
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
| | - Julia Lorenzo
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Carles Arús
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (P.A.-T.); (J.L.); (A.P.C.); (C.A.)
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Daniel Ruiz-Molina
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
| | - Fernando Novio
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
- Departament de Química, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
43
|
Shimozaki K, Nakayama I, Hirota T, Yamaguchi K. Current Strategy to Treat Immunogenic Gastrointestinal Cancers: Perspectives for a New Era. Cells 2023; 12:1049. [PMID: 37048122 PMCID: PMC10093684 DOI: 10.3390/cells12071049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/15/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Since pembrolizumab, an anti-programmed death-1 (PD-1) antibody, showed a dramatic response to immunogenic cancers with microsatellite instability-high (MSI-H) and/or deficient mismatch repair (dMMR) in the pilot clinical trial KEYNOTE-016, subsequent studies have confirmed durable responses of anti-PD-1 inhibitors for MSI-H/dMMR solid tumors. As immunotherapy is described as a "game changer," the therapeutic landscape for MSI-H/dMMR solid tumors including gastrointestinal cancers has changed considerably in the last decade. An MSI/MMR status has been established as the predictive biomarker for immune checkpoint blockades, playing an indispensable role in the clinical practice of patients with MSI-H/dMMR tumors. Immunotherapy is also now investigated for locally advanced MSI-H/dMMR gastrointestinal cancers. Despite this great success, a few populations with MSI-H/dMMR gastrointestinal cancers do not respond to immunotherapy, possibly due to the existence of intrinsic or acquired resistance mechanisms. Clarifying the underlying mechanisms of resistance remains a future task, whereas attempts to overcome resistance and improve the efficacy of immunotherapy are currently ongoing. Herein, we review recent clinical trials with special attention to MSI-H/dMMR gastrointestinal cancers together with basic/translational findings, which provide their rationale, and discuss perspectives for the further therapeutic development of treatment in this field.
Collapse
Affiliation(s)
- Keitaro Shimozaki
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Izuma Nakayama
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan
| | - Toru Hirota
- Department of Experimental Pathology, Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan
| |
Collapse
|
44
|
Prashantha K, Krishnappa A, Muthappa M. 3D bioprinting of gastrointestinal cancer models: A comprehensive review on processing, properties, and therapeutic implications. Biointerphases 2023; 18:020801. [PMID: 36963961 DOI: 10.1116/6.0002372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023] Open
Abstract
Gastrointestinal tract (GIT) malignancies are an important public health problem considering the increased incidence in recent years and the high morbidity and mortality associated with it. GIT malignancies constitute 26% of the global cancer incidence burden and 35% of all cancer-related deaths. Gastrointestinal cancers are complex and heterogenous diseases caused by the interplay of genetic and environmental factors. The tumor microenvironment (TME) of gastrointestinal tract carcinomas is dynamic and complex; it cannot be recapitulated in the basic two-dimensional cell culture systems. In contrast, three-dimensional (3D) in vitro models can mimic the TME more closely, enabling an improved understanding of the microenvironmental cues involved in the various stages of cancer initiation, progression, and metastasis. However, the heterogeneity of the TME is incompletely reproduced in these 3D culture models, as they fail to regulate the orientation and interaction of various cell types in a complex architecture. To emulate the TME, 3D bioprinting has emerged as a useful technique to engineer cancer tissue models. Bioprinted cancer tissue models can potentially recapitulate cancer pathology and increase drug resistance in an organ-mimicking 3D environment. In this review, we describe the 3D bioprinting methods, bioinks, characterization of 3D bioprinted constructs, and their application in developing gastrointestinal tumor models that integrate their microenvironment with different cell types and substrates, as well as bioprinting modalities and their application in therapy and drug screening. We review prominent studies on the 3D bioprinted esophageal, hepatobiliary, and colorectal cancer models. In addition, this review provides a comprehensive understanding of the cancer microenvironment in printed tumor models, highlights current challenges with respect to their clinical translation, and summarizes future perspectives.
Collapse
Affiliation(s)
- Kalappa Prashantha
- Centre for Research and Innovation, Adichunchanagiri School of Natural Sciences, Adichunchanagiri University, BGSIT, B.G. Nagara, Mandya District 571448, Karnataka, India
| | - Amita Krishnappa
- Department of Pathology, Adichunchanagiri Institute of Medicinal Sciences Adichunchanagiri University, B.G. Nagara, Mandya District 571448, Karnataka, India
| | - Malini Muthappa
- Department of Physiology, Adichunchanagiri Institute of Medicinal Sciences Adichunchanagiri University, B.G. Nagara, Mandya District 571448, Karnataka, India
| |
Collapse
|
45
|
Gielecińska A, Kciuk M, Mujwar S, Celik I, Kołat D, Kałuzińska-Kołat Ż, Kontek R. Substances of Natural Origin in Medicine: Plants vs. Cancer. Cells 2023; 12:986. [PMID: 37048059 PMCID: PMC10092955 DOI: 10.3390/cells12070986] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Continuous monitoring of the population's health is the main method of learning about disease prevalence. National and international data draw attention to the persistently high rates of cancer incidence. This necessitates the intensification of efforts aimed at developing new, more effective chemotherapeutic and chemopreventive drugs. Plants represent an invaluable source of natural substances with versatile medicinal properties. Multidirectional activities exhibited by natural substances and their ability to modulate key signaling pathways, mainly related to cancer cell death, make these substances an important research direction. This review summarizes the information regarding plant-derived chemotherapeutic drugs, including their mechanisms of action, with a special focus on selected anti-cancer drugs (paclitaxel, irinotecan) approved in clinical practice. It also presents promising plant-based drug candidates currently being tested in clinical and preclinical trials (betulinic acid, resveratrol, and roburic acid).
Collapse
Affiliation(s)
- Adrianna Gielecińska
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| | - Mateusz Kciuk
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Turkey
| | - Damian Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
46
|
Dhara V, Shetty SS, de Arruda JAA, Silva TA, Russo RC, Shetty NJ, Pidaparthi M, Wollenberg B, Rao VUS, Gopinath TPS. Decoding the influence of the immune system and immunotherapy targets on carcinomas: A hidden prism in oral cancer therapy. Dis Mon 2023; 69:101353. [PMID: 35311656 DOI: 10.1016/j.disamonth.2022.101353] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In recent decades, understanding tumorigenesis and the complex interaction between the host and the immune system has been the pillar for significant advances in anticancer therapy. Conventional anticancer therapy (e.g., cut, burn, and cytotoxic drugs) involves multiple targeting of tumor cells. However, the tumor tissue microenvironment can present a dysregulated, stimulating, or subverted immune response which, in turn, reveals pro-tumor activities favoring tumor expansion and progression. Recently, new potential targets have been identified based on immunomodulatory therapies, which are crafted to re-establish the host anti-tumoral immune response. Clinicians should fully understand the intricate interactions between carcinogens, the tumor milieu, the immune system, and traditional anticancer therapies in order to progress and to overcome the refractory/recurrent challenges and morbidity of the disease. Thus, in this article, we highlight the complex milieu of the oral cancer immune response, pointing out potential therapeutic immunotargets for oral squamous cell carcinomas. The impact of traditional anticancer therapy on the immune system is also outlined.
Collapse
Affiliation(s)
- Vasantha Dhara
- Consultant Maxillofacial Surgeon, Hyderabad, Telangana, India
| | - Sameep S Shetty
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, Manipal Academy of Higher Education, A constituent of MAHE, Manipal, Karnataka, India.
| | - José Alcides Almeida de Arruda
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Tarcília Aparecida Silva
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Neetha J Shetty
- Department of Periodontology, Manipal College of Dental Sciences, Mangalore, Manipal Academy of Higher Education, A constituent of MAHE, Mangalore, Karnataka, India
| | | | - Barbara Wollenberg
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Klinikum rechts der Isar der TU München, Ismaningerstraße 22, 81675 München, Germany
| | - Vishal U S Rao
- Department of Head and Neck Surgical Oncology, HealthCare Global Enterprises Ltd., Bangalore, Karnataka, India
| | - Thilak P S Gopinath
- Nitte (Deemed to be University) , AB Shetty Memorial Institute of Dental Sciences (ABSMIDS) , Department of Oral and Maxillofacial Surgery, Mangalore, India
| |
Collapse
|
47
|
Wang AX, Ong XJ, D’Souza C, Neeson PJ, Zhu JJ. Combining chemotherapy with CAR-T cell therapy in treating solid tumors. Front Immunol 2023; 14:1140541. [PMID: 36949946 PMCID: PMC10026332 DOI: 10.3389/fimmu.2023.1140541] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Chemotherapy has long been a standard treatment for a wide range of malignancies, where patients typically undergo multiple rounds of chemotherapy regimens to control tumor growth. In the clinic, the chemotherapy drugs cyclophosphamide and fludarabine are commonly used prior to Chimeric Antigen Receptor T (CAR-T) cell therapy to lymphodeplete and improve CAR-T cell engraftment. In this review, we discuss the use of chemotherapy in combination with CAR-T cell therapy. We also show that chemotherapy can deplete immunosuppressive cells, promote a pro-inflammatory tumor microenvironment, disrupt tumor stroma, and improve CAR-T cell recruitment to the tumor. Although the combination of chemotherapy plus CAR-T cell therapy is promising, certain aspects of chemotherapy also pose a challenge. In addition, the combined therapeutic effect may be heavily dependent on the dose and the treatment schedule. Thus, we also discussed the obstacles to effective clinical outcomes of the combination therapy.
Collapse
Affiliation(s)
- Arthur Xuan Wang
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Xiao Jing Ong
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Criselle D’Souza
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, VIC, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, VIC, Australia
| | - Joe Jiang Zhu
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Joe Jiang Zhu,
| |
Collapse
|
48
|
Khadela A, Shah Y, Mistry P, Bodiwala K, CB A. Immunomodulatory Therapy in Head and Neck Squamous Cell Carcinoma: Recent Advances and Clinical Prospects. Technol Cancer Res Treat 2023; 22:15330338221150559. [PMID: 36683526 PMCID: PMC9893386 DOI: 10.1177/15330338221150559] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The immune system plays a significant role in the development, invasion, progression, and metastasis of head and neck cancer. Over the last decade, the emergence of immunotherapy has irreversibly altered the paradigm of cancer treatment. The current treatment modalities for head and neck squamous cell carcinoma (HNSCC) include surgery, radiotherapy, and adjuvant or neoadjuvant chemotherapy which has failed to provide satisfactory clinical outcomes. To encounter this, there is a need for a novel or targeted therapy such as immunological targets along with conventional treatment strategy for optimal therapeutic outcomes. The immune system can contribute to promoting metastasis, angiogenesis, and growth by exploiting the tumor's influence on the microenvironment. Immunological targets have been found effective in recent clinical studies and have shown promising results. This review outlines the important immunological targets and the medications acting on them that have already been explored, are currently under clinical trials and are further being targeted.
Collapse
Affiliation(s)
- Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Yesha Shah
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Priya Mistry
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Kunjan Bodiwala
- Department of Pharmaceutical chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Avinash CB
- Medical Oncologist, ClearMedi Radiant Hospital, Mysore, India
| |
Collapse
|
49
|
Chang X, Bian M, Liu L, Yang J, Yang Z, Wang Z, Lu Y, Liu W. Induction of immunogenic cell death by novel platinum-based anticancer agents. Pharmacol Res 2023; 187:106556. [PMID: 36403722 DOI: 10.1016/j.phrs.2022.106556] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/02/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022]
Abstract
Traditional platinum-based anticancer drugs, led by cisplatin, play an important role in chemotherapy. However, the development of platinum compounds is limited due to serious toxicity and side effects. In recent years, studies have showed that immunogenic cell death (ICD) may be one of the potential action mechanisms of classical platinum drugs, such as oxaliplatin. This strategy combining chemotherapy and immunotherapy can effectively utilize the body's immune system to help platinum compounds to fight against tumors, and the dose can be appropriately reduced to limit toxic side effects. The induction of ICD by platinum compounds has become a research hotspot and one of the future development directions of metal drugs. Here, the progress of platinum compounds were collected and comprehensively summarized, their capacity of ICD induction and mechanism of action are exposed, providing reference for the design and synthesis of new anticancer platinum ICD inducers.
Collapse
Affiliation(s)
- Xingyu Chang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Mianli Bian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Lijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jiaqi Yang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhibin Yang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhaoran Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
50
|
Stipp MC, Corso CR, Acco A. Impacts of COVID-19 in Breast Cancer: From Molecular Mechanism to the Treatment Approach. Curr Pharm Biotechnol 2023; 24:238-252. [PMID: 35593354 DOI: 10.2174/1389201023666220421133311] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/17/2021] [Accepted: 03/03/2022] [Indexed: 11/22/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has already infected more than 272 million people, resulting in 5.3 million deaths worldwide from COVID-19. Breast tumors are considered the world's most commonly diagnosed cancer. Both breast cancer and COVID-19 share common pathogenic features, represented by inflammatory mediators and the potential of SARS-CoV-2 replication in metastatic cancer cells. This may intensify viral load in patients, thereby triggering severe COVID-19 complications. Thus, cancer patients have a high risk of developing severe COVID-19 with SARS-CoV-2 infection and a higher rate of complications and death than non-cancer patients. The present review discusses common mechanisms between COVID-19 and breast cancer and the particular susceptibility to COVID-19 in breast cancer patients. We describe the effects of chemotherapeutic agents that are used against this cancer, which should be considered from the perspective of susceptibility to SARS-CoV-2 infection and risk of developing severe events. We also present potential drug interactions between chemotherapies that are used to treat breast cancer and drugs that are applied for COVID-19. The drugs that are identified as having the most interactions are doxorubicin and azithromycin. Both drugs can interact with each other and with other drugs, which likely requires additional drug monitoring and changes in drug dosage and timing of administration. Further clinical and observational studies involving breast cancer patients who acquire COVID-19 are needed to define the best therapeutic approach when considering the course of both diseases.
Collapse
Affiliation(s)
- Maria Carolina Stipp
- Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | | | - Alexandra Acco
- Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| |
Collapse
|