1
|
Kret ZS, Sweder RJ, Pollock R, Tinoco G. Potential Mechanisms for Immunotherapy Resistance in Adult Soft-Tissue Sarcoma. Target Oncol 2025; 20:485-502. [PMID: 40289241 DOI: 10.1007/s11523-025-01145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Soft-tissue sarcomas represent a diverse group of rare malignancies originating from mesenchymal tissue, accounting for less than 1% of adult cancers in the USA. With over 13,000 new cases and around 5350 deaths annually, patients with metastatic soft-tissue sarcomas face limited therapeutic options and an estimated median overall survival of 18 months. While immunotherapy has demonstrated effectiveness in several cancers, its application in soft-tissue sarcomas remains challenging owing to the tumors' largely "cold" immunological environment, characterized by low levels of tumor-infiltrating lymphocytes and a lack of soft-tissue sarcoma-specific biomarkers. This review examines potential mechanisms underlying immunotherapy resistance in soft-tissue sarcomas, including the complex interplay between innate and adaptive immunity, the tumor microenvironment, and the role of immune-related genes. Despite preliminary findings suggesting correlations between immune profiles and histological subtypes, consistent biomarkers for predicting immunotherapeutic responses across soft-tissue sarcoma types are absent. Emerging strategies focus on converting "cold" tumors to "hot" tumors, enhancing their susceptibility to immunologic activation. While research is ongoing, personalized treatment approaches may offer hope for overcoming the inherent heterogeneity and resistance seen in soft-tissue sarcomas, ultimately aiming to improve outcomes for affected patients.
Collapse
Affiliation(s)
- Zaina S Kret
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ryan J Sweder
- The Ohio State University College of Arts and Sciences and College of Medicine, Columbus, OH, USA
| | - Raphael Pollock
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 1800 Cannon Drive, 1240 Lincoln Tower, Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
Osama M, Kocherry C, Ullah F, Ubaid S, Ubaid M, Ullah U, Nawaz AB, Qasem HM, Odat RM, Farhan M, Ahmed R. Odontogenic carcinosarcoma: a comprehensive review of clinical and therapeutic insights. FRONTIERS IN ORAL HEALTH 2025; 6:1544921. [PMID: 40336633 PMCID: PMC12055786 DOI: 10.3389/froh.2025.1544921] [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: 12/13/2024] [Accepted: 03/26/2025] [Indexed: 05/09/2025] Open
Abstract
Malignant odontogenic tumors are rare, accounting for only 1%-6.1% of all odontogenic tumors. Among them, odontogenic carcinosarcoma (OCS) is an exceptionally rare and aggressive malignant neoplasm originating from dental tissues. First recognized by the World Health Organization (WHO) in 1992, OCS is characterized by high-grade biphasic malignant epithelial and mesenchymal components, contributing to its aggressive clinical behavior. OCS often presents with nonspecific symptoms such as pain, swelling, and loosening of teeth, which complicate early diagnosis. Its rarity adds to the diagnostic challenges, frequently leading to delays in identification. Histopathological evaluation remains the cornerstone for accurate diagnosis, distinguishing OCS from other odontogenic tumors through features like epithelial nuclear pleomorphism, mitotic activity, and mesenchymal sarcomatous differentiation. Management typically involves surgical resection with clear margins, while adjuvant therapies such as chemotherapy and radiation are considered in select cases. Recent advancements in molecular oncology and surgical techniques, including robotic-assisted procedures and 3D-printed reconstructive aids, offer promising avenues for improving patient outcomes. A multidisciplinary approach and ongoing research are essential to enhance diagnostic accuracy, refine treatment protocols, and improve the prognosis for patients affected by this rare malignancy. The primary objective of this review is to consolidate current knowledge on OCS, focusing on its diagnostic complexities, treatment strategies, and potential emerging therapies.
Collapse
Affiliation(s)
- Muhammad Osama
- Internal Medicine, Khyber Medical College, Peshawar, Pakistan
| | - Cyril Kocherry
- School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Farid Ullah
- Internal Medicine, Khyber Teaching Hospital, Peshawar, Pakistan
| | - Safiyyah Ubaid
- Department of Biochemistry, Khyber Girls Medical College, Peshawar, Pakistan
| | - Maryam Ubaid
- Public Health, Khyber Medical University, Peshawar, Pakistan
| | - Ubaid Ullah
- Department of Biochemistry, Kabir Medical College, Peshawar, Pakistan
| | | | - Hanan M. Qasem
- Faculty of Dentistry, Jordan University of Science and Technology, Irbid, Jordan
| | - Ramez M. Odat
- Faculty of Medicine, Jordan University of Science & Technology, Irbid, Jordan
| | - Muzammil Farhan
- Department of Cardiology, Imperial College London, London, United Kingdom
| | - Raheel Ahmed
- Department of Cardiology, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Martin-Broto J, Moura DS, Hindi N. Which sarcoma requires PD1/PDL1 inhibitors, and what should be the best scheme? Present status and next steps. Curr Opin Oncol 2025:00001622-990000000-00254. [PMID: 40421971 DOI: 10.1097/cco.0000000000001149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
PURPOSE OF REVIEW The introduction of immune checkpoint inhibitors (ICI) in advanced sarcoma has been largely disappointing due to their "cold" tumor microenvironment, characterized by low tumor mutational burden, scarce CD8+ T-cell infiltration, and minimal expression of PD-1/PD-L1. However, recent findings highlight several scenarios in which immune checkpoint blockade exhibits clinical efficacy. RECENT FINDINGS ICIs have shown durable efficacy in specific sarcoma subtypes, such as alveolar soft part sarcoma (ASPS), with objective response rates (ORR) exceeding 35% and 50%, in monotherapy or in combination, respectively. Doxorubicin-based regimens plus ICIs have yielded notorious and higher ORRs in the most common sarcoma subtypes, than historical chemotherapy data. Neoadjuvant radiation therapy combined with ICIs has significantly improved disease-free survival in localized selected soft tissue sarcomas. SUMMARY Immunotherapy targeting immune checkpoints in sarcomas is evolving, with recent findings highlighting its potential. Single-arm trials underscore the efficacy of ICIs in rare sarcomas, exemplified by the FDA approval of atezolizumab for ASPS. Combination strategies are proving more effective than chemotherapy alone, with ongoing comparative studies assessing chemo-immunotherapy in both metastatic and localized sarcomas. Advances in predictive biomarkers could expand the clinical use of ICIs.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital; University Hospital General de Villalba, and Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM)
| | - David S Moura
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM) and University Hospital General de Villalba, Madrid, Spain
| | - Nadia Hindi
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital; University Hospital General de Villalba, and Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM)
| |
Collapse
|
4
|
Benton A, Liu B, Gartenhaus LE, Hanna JA. Genomic landscape and preclinical models of angiosarcoma. Mol Oncol 2025; 19:965-983. [PMID: 39367667 PMCID: PMC11977660 DOI: 10.1002/1878-0261.13744] [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: 07/01/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
Angiosarcoma is a cancer that develops in blood or lymphatic vessels that presents a significant clinical challenge due to its rarity and aggressive features. Clinical outcomes have not improved in decades, highlighting a need for innovative therapeutic strategies to treat the disease. Genetically, angiosarcomas exhibit high heterogeneity and complexity with many recurrent mutations. However, recent studies have identified some common features within anatomic and molecular subgroups. To identify potential therapeutic vulnerabilities, it is essential to understand and integrate the mutational landscape of angiosarcoma with the models that exist to study the disease. In this review, we will summarize the insights gained from reported genomic alterations in molecular and anatomic subtypes of angiosarcoma, discuss several potential actionable targets, and highlight the preclinical disease models available in the field.
Collapse
Affiliation(s)
- Annaleigh Benton
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
- Purdue University Institute for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| | - Bozhi Liu
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
- Purdue University Institute for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| | - Lauren E. Gartenhaus
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
- Purdue University Institute for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| | - Jason A. Hanna
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
- Purdue University Institute for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
5
|
Chen TWW. Angiosarcoma: Role of Immunotherapy. Curr Treat Options Oncol 2025; 26:242-250. [PMID: 40056281 DOI: 10.1007/s11864-025-01307-7] [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] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
OPINION STATEMENT Recent clinical trials have investigated immune checkpoint inhibitors (ICIs) alone, in combination with tyrosine kinase inhibitors (TKIs), or with chemotherapy in angiosarcoma, yielding mixed results across subtypes. Single-agent ICI therapy, such as cemiplimab (ORR 27.8%), has shown responses primarily in UV- and radiation-associated angiosarcomas, likely due to their higher tumor mutation burden (TMB), while dual ICI therapy (SWOG S1609, ORR 25%) suggests potential benefit but remains limited in cutaneous disease. ICI-TKI combinations, such as cabozantinib plus nivolumab (Alliance A091902, second-line, ORR 59%), demonstrated significant efficacy in both cutaneous and non-cutaneous angiosarcomas, suggesting anti-angiogenic therapy may enhance ICI responses in tumors historically resistant to checkpoint blockade. ICI-chemotherapy combinations have been less consistent. The Alliance A091902 first-line trial (paclitaxel ± nivolumab) found no overall PFS benefit, though scalp/face angiosarcoma patients appeared to fare better, raising the question of whether ICI alone might be equally effective in this subset. The South Korean paclitaxel + avelumab trial (ORR 50%) showed promising response rates, but the lack of detailed subgroup analysis limits interpretation. Other chemotherapy-ICI combinations, such as doxorubicin plus pembrolizumab, have shown isolated responses but require further study in larger cohorts. Moving forward, better biomarkers are critical for identifying which patients benefit most from ICIs, and while TKI-ICI combinations appear to hold the most promise, chemotherapy-ICI strategies need further refinement to optimize sequencing and patient selection in angiosarcoma treatment.
Collapse
Affiliation(s)
- Tom Wei-Wu Chen
- Department of Oncology and Center of Excellence for Sarcoma, National Taiwan University Hospital, 7 Chung Shan South Rd, Taipei, Taiwan, 100.
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
| |
Collapse
|
6
|
Adil ANK, Hassan M, Khan M, Rahman B, Raza SS, Varrassi G. Clear Cell Sarcoma of the Knee: A Case Report of Diagnostic Challenges, Rapid Metastatic Spread, and Treatment Dilemmas. Cureus 2025; 17:e80996. [PMID: 40260360 PMCID: PMC12011346 DOI: 10.7759/cureus.80996] [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: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/23/2025] Open
Abstract
Clear cell sarcoma (CCS) is a rare, aggressive soft tissue sarcoma with a strong predilection for the extremities. It is often misdiagnosed as melanoma. Despite surgical resection, CCS carries a high risk of early metastasis, most commonly to the lungs, bones, and lymph nodes, with limited effective systemic therapies available. We present the case of a 48-year-old man with CCS of the knee, initially presenting as a localized disease but rapidly progressing to widespread metastasis within three months. The patient underwent radical resection with knee arthroplasty and adjuvant radiotherapy, followed by systemic therapy with cabozantinib, ipilimumab, and nivolumab. Despite aggressive treatment, the disease progressed, leading to recurrent pleural effusions, respiratory failure, and, ultimately, the patient's demise within 10 months of diagnosis. This case underscores the highly aggressive nature of CCS and the challenges associated with its management. The rapid metastatic spread despite multimodal therapy highlights the need for improved early detection strategies and more effective systemic treatments, including novel targeted therapies and immunotherapy combinations.
Collapse
Affiliation(s)
| | | | - Muhammad Khan
- Internal Medicine, University of Kansas Medical Center, Kansas City, USA
| | - Bakht Rahman
- Internal Medicine, Community Medical Center, Fresno, USA
| | - Syed S Raza
- Internal Medicine, Community Medical Center, Fresno, USA
- Physiology, Khyber Medical College/Teaching Hospital, Peshawar, PAK
| | | |
Collapse
|
7
|
Cao Y, Wang W, Xu H, Yi H, Gao Y, Wan M, Wang M, Chen T, Chen Y, Chi Y, Wei S, Jin S, Bai M, Li X, Gao Y, Niu X, Liu Y. Efficacy of immune checkpoint inhibitors in the treatment of soft tissue sarcoma: A systematic review and meta-analysis of clinical trials. Int Immunopharmacol 2025; 148:114070. [PMID: 39826454 DOI: 10.1016/j.intimp.2025.114070] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Soft tissue sarcomas (STS) are a heterogeneous group of tumors with diverse clinical and molecular characteristics, characterized by limited treatment options and poor prognosis. Immune checkpoint inhibitors (ICIs) have emerged as promising therapies for STS, yet comprehensive evaluations of their efficacy, especially in combination with other treatments, are scarce. METHODS We conducted a systematic review and meta-analysis of clinical trials on ICIs in STS treatment, sourced from PubMed, Embase, and the Cochrane Central Register of Controlled Trials up to May 31, 2024. The studies included both monotherapy and combination therapies with ICIs. We assessed the methodological quality using the Cochrane Risk of Bias 2 tool and the Methodological Index for Non-Randomized Studies. Data synthesis involved random-effects meta-analysis to determine pooled proportions and 95% confidence intervals (CIs) for objective response rates (ORR), disease control rates (DCR), and high-grade treatment-related adverse events (TRAEs). RESULTS The analysis included 38 studies with 1349 patients covering 24 STS subtypes. The overall ORR was 16% (95% CI 0.12-0.21), DCR was 64% (95% CI 0.57-0.70), and the rate of Grade 3-5 TRAEs was 19% (95% CI 0.13-0.27). Treatments combining ICIs with tyrosine kinase inhibitors (TKIs) showed the highest efficacy (ORR 28%, 95% CI 0.18-0.40), albeit with increased adverse events. ORRs in first-line treatments were substantially higher (28%) compared to second-line treatments or beyond (11%). Subtypes like alveolar soft part sarcoma (ASPS), angiosarcoma (AS), and epithelioid sarcoma (ES) exhibited favorable responses exceeding 30%. CONCLUSIONS This systematic review and meta-analysis indicate that ICIs, particularly when combined with TKIs, provide substantial therapeutic benefits in treating STS, significantly enhancing response rates in specific subtypes such as ASPS and AS. The results underscore the transformative potential of ICIs in STS treatment strategies. However, the variability across subtypes and treatment lines emphasizes the need for further randomized controlled trials to refine and personalize therapeutic approaches, ensuring optimal outcomes for patients with these diverse malignancies.
Collapse
Affiliation(s)
- Yang Cao
- 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, China
| | - Wei Wang
- 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, China
| | - Hairong Xu
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No.31 Xin Jie Kou East Street, Xi Cheng District, Beijing 100035, China
| | - Hang Yi
- Department of Thoracic 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
| | - Yinyan Gao
- Department of Epidemiology and Biostatistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Mingzhong Wan
- Shantou University Medical College, Shantou 515041, China
| | - Mingzhao Wang
- 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, China
| | - Tong Chen
- 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, China
| | - Yanchao Chen
- 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, China
| | - Yihebali Chi
- 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, China
| | - Shuqing Wei
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, General Internal Medicine, Taiyuan, Shanxi 030013, China
| | - Shi Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Ming Bai
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Xin Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Yibo Gao
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China.
| | - Xiaohui Niu
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No.31 Xin Jie Kou East Street, Xi Cheng District, Beijing 100035, China.
| | - Yutao Liu
- 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, China.
| |
Collapse
|
8
|
Liu H, Hao Q, Wang X, Cheng M, Qiu F, Zhou B. Efficacy and safety of the combination of anlotinib and envafolimab in the treatment of unresectable or metastatic liposarcoma: findings from a single-center retrospective study. Front Oncol 2025; 14:1502945. [PMID: 39868378 PMCID: PMC11757892 DOI: 10.3389/fonc.2024.1502945] [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: 09/27/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Objective To evaluate the efficacy and safety of anlotinib combined with envafolimab in the treatment of unresectable or metastatic liposarcoma. Methods This single-center, retrospective study enrolled 15 patients with unresectable or metastatic liposarcoma, who were treated at the Retroperitoneal Tumor Surgery Research Center of Qingdao University Affiliated Hospital between April 2022 and November 2023. The treatment regimen consisted of anlotinib combined with envafolimab. Treatment efficacy was evaluated using the Response Evaluation Criteria in Solid Tumors version 1.1. Treatment-related adverse events (TRAEs) were assessed using Common Terminology Criteria for Adverse Events version 5.0. Results A total of 15 patients with unresectable or metastatic liposarcoma were included; among them, seven were male (46.7%) and eight were female (53.3%), with a median age of 55 years. The pathological subtype distribution was as follows: three (20.0%) patients with well-differentiated liposarcoma, 11 (73.3%) patients with dedifferentiated liposarcoma, and one (6.7%) patient with myxoid liposarcoma. At 12 weeks post-diagnosis, none of the patients achieved a complete response. The objective response rate was 6.7%, with one patient (6.7%) achieving a partial response. Disease stability was observed in 10 (66.6%) patients, which corresponded to a disease control rate of 73.3%. Disease progression occurred in four (26.7%) patients. The median follow-up time was 16.9 months and the median progression-free survival time was 14.2 months. Seven patients experienced TRAEs, of whom three (42.2%) had grade 3-4 TRAEs. The most common TRAEs were liver function abnormalities, hypertension, and fatigue. Conclusion Anlotinib combined with envafolimab demonstrates promising efficacy and manageable safety in treating unresectable or metastatic liposarcoma.
Collapse
Affiliation(s)
- Hongliang Liu
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qisheng Hao
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xi Wang
- Department of Oncology, Women and Children’s Hospital Affiliated to Qingdao University, Qingdao, China
| | - Mengxing Cheng
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fabo Qiu
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Cao Z, Quazi S, Arora S, Osellame LD, Burvenich IJ, Janes PW, Scott AM. Cancer-associated fibroblasts as therapeutic targets for cancer: advances, challenges, and future prospects. J Biomed Sci 2025; 32:7. [PMID: 39780187 PMCID: PMC11715488 DOI: 10.1186/s12929-024-01099-2] [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: 04/05/2024] [Accepted: 11/09/2024] [Indexed: 01/11/2025] Open
Abstract
Research into cancer treatment has been mainly focused on developing therapies to directly target cancer cells. Over the past decade, extensive studies have revealed critical roles of the tumour microenvironment (TME) in cancer initiation, progression, and drug resistance. Notably, cancer-associated fibroblasts (CAFs) have emerged as one of the primary contributors in shaping TME, creating a favourable environment for cancer development. Many preclinical studies have identified promising targets on CAFs, demonstrating remarkable efficacy of some CAF-targeted treatments in preclinical models. Encouraged by these compelling findings, therapeutic strategies have now advanced into clinical evaluation. We aim to provide a comprehensive review of relevant subjects on CAFs, including CAF-related markers and targets, their multifaceted roles, and current landscape of ongoing clinical trials. This knowledge can guide future research on CAFs and advocate for clinical investigations targeting CAFs.
Collapse
Affiliation(s)
- Zhipeng Cao
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia.
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, 3084, Australia.
| | - Sadia Quazi
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Sakshi Arora
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Laura D Osellame
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Ingrid J Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Peter W Janes
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia.
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, 3084, Australia.
- Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
10
|
Zhao S, Du H, Zhang Z, Yang J, Zhou Y, Xiao G, Ma H, Lan C, Liang J, Yang K, Wen L. SMARCB1/INI-1-Deficient sinonasal carcinoma demonstrates a poor prognosis but favorable clinical outcomes after PD-1/PD-L1 inhibitor therapy: A case series. Sci Prog 2025; 108:368504251315075. [PMID: 39886754 PMCID: PMC11783480 DOI: 10.1177/00368504251315075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Despite advances in multimodal cancer therapy, such as combining radical surgery with high-intensity chemoradiotherapy, for SMARCB1/INI-1-deficient sinonasal carcinoma (SDSC), the prognosis of patients remains poor. Immunotherapy is gaining increasing popularity as a novel treatment strategy for patients with SMARCB1/INI-1-deficient tumors. Herein, we report on the management of three patients with SDSC who received PD-1/PD-L1 inhibitor therapy as a part of multimodal therapy based on surgery and chemoradiotherapy. All three patients survived and demonstrated good clinical remission and disease control. To our knowledge, this is the first case series reporting the use of immunotherapy to improve clinical outcomes in neoadjuvant, adjuvant, and late first-line stages of treatment in patients with SDSC. Furthermore, we reviewed the relevant literature and further explored the correlation between SMARCB1/INI-1 deletion and immunotherapy.
Collapse
Affiliation(s)
- Shuhan Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Hao Du
- Department of Neurosurgery, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanjie Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Jinsong Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - You Zhou
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guixiang Xiao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Ma
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caini Lan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Jinzi Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Lu Wen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| |
Collapse
|
11
|
Deshmukh S, Kelly C, Tinoco G. IDH1/2 Mutations in Cancer: Unifying Insights and Unlocking Therapeutic Potential for Chondrosarcoma. Target Oncol 2025; 20:13-25. [PMID: 39546097 DOI: 10.1007/s11523-024-01115-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
Chondrosarcomas, a rare form of bone sarcomas with multiple subtypes, pose a pressing clinical challenge for patients with advanced or metastatic disease. The lack of US Food and Drug Administration (FDA)-approved medications underscores the urgent need for further research and development in this area. Patients and their families face challenges as there are no systemic therapeutic options available with substantial effectiveness. A significant number (50-80%) of chondrosarcomas have a mutation in the isocitrate dehydrogenase (IDH) genes. This review focuses on IDH-mediated pathogenesis and recent pharmacological advances with novel IDH inhibitors, explores their potential therapeutic value, and proposes potential future avenues for clinical trials combining IDH inhibitors with other systemic agents for chondrosarcomas.
Collapse
Affiliation(s)
- Shriya Deshmukh
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ciara Kelly
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabriel Tinoco
- Division of Medical Oncology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
12
|
Hayes AJ, Nixon IF, Strauss DC, Seddon BM, Desai A, Benson C, Judson IR, Dangoor A. UK guidelines for the management of soft tissue sarcomas. Br J Cancer 2025; 132:11-31. [PMID: 38734790 PMCID: PMC11724041 DOI: 10.1038/s41416-024-02674-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 05/13/2024] Open
Abstract
Soft tissue sarcomas (STS) are rare tumours arising in mesenchymal tissues and can occur almost anywhere in the body. Their rarity, and the heterogeneity of subtype and location, means that developing evidence-based guidelines is complicated by the limitations of the data available. This makes it more important that STS are managed by expert multidisciplinary teams, to ensure consistent and optimal treatment, recruitment to clinical trials, and the ongoing accumulation of further data and knowledge. The development of appropriate guidance, by an experienced panel referring to the evidence available, is therefore a useful foundation on which to build progress in the field. These guidelines are an update of the previous versions published in 2010 and 2016 [1, 2]. The original guidelines were drawn up by a panel of UK sarcoma specialists convened under the auspices of the British Sarcoma Group (BSG) and were intended to provide a framework for the multidisciplinary care of patients with soft tissue sarcomas. This iteration of the guidance, as well as updating the general multidisciplinary management of soft tissue sarcoma, includes specific sections relating to the management of sarcomas at defined anatomical sites: gynaecological sarcomas, retroperitoneal sarcomas, breast sarcomas, and skin sarcomas. These are generally managed collaboratively by site specific multidisciplinary teams linked to the regional sarcoma specialist team, as stipulated in the recently published sarcoma service specification [3]. In the UK, any patient with a suspected soft tissue sarcoma should be referred to a specialist regional soft tissues sarcoma service, to be managed by a specialist sarcoma multidisciplinary team. Once the diagnosis has been confirmed using appropriate imaging and a tissue biopsy, the main modality of management is usually surgical excision performed by a specialist surgeon, combined with pre- or post-operative radiotherapy for tumours at higher risk for local recurrence. Systemic anti-cancer therapy (SACT) may be utilised in cases where the histological subtype is considered more sensitive to systemic treatment. Regular follow-up is recommended to assess local control, development of metastatic disease, and any late effects of treatment.
Collapse
Affiliation(s)
- Andrew J Hayes
- The Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK.
- The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Ioanna F Nixon
- Department of Clinical Oncology, The Beatson West of Scotland Cancer Center, Glasgow, G12 0YN, UK
| | - Dirk C Strauss
- The Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Beatrice M Seddon
- Department of Medical Oncology, University College London Hospital NHS Foundation Trust, London, NW1 2BU, UK
| | - Anant Desai
- The Midlands Abdominal and Retroperitoneal Sarcoma Unit, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Charlotte Benson
- The Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Ian R Judson
- The Institute of Cancer Research, London, SM2 5NG, UK
| | - Adam Dangoor
- Department of Medical Oncology, University Hospitals Bristol & Weston NHS Foundation Trust, Bristol, BS1 3NU, UK
| |
Collapse
|
13
|
Palmerini E, Lopez Pousa A, Grignani G, Redondo A, Hindi N, Provenzano S, Sebio A, Lopez Martin JA, Valverde C, Martinez Trufero J, Gutierrez A, de Alava E, Aparisi Gomez MP, D'Ambrosio L, Collini P, Bazzocchi A, Moura DS, Ibrahim T, Stacchiotti S, Broto JM. Nivolumab and sunitinib in patients with advanced bone sarcomas: A multicenter, single-arm, phase 2 trial. Cancer 2025; 131:e35628. [PMID: 39540661 DOI: 10.1002/cncr.35628] [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: 07/20/2024] [Revised: 08/31/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Herein, we present the results of the phase 2 IMMUNOSARC study (NCT03277924), investigating sunitinib and nivolumab in adult patients with advanced bone sarcomas (BS). METHODS Progressing patients with a diagnosis of BS were eligible. Treatment was comprised of sunitinib (37.5 mg/day on days 1-14, 25 mg/day afterword) plus nivolumab (3 mg/kg every 2 weeks). Primary end point was progression-free survival rate (PFSR) at 6 months based on central radiology review. Secondary end points were overall survival (OS), overall response rate (ORR) by Response Evaluation Criteria in Solid Tumors (RECIST) v1.1, and safety. RESULTS A total of 46 patients were screened, 40 patients entered the study, and 38 underwent central radiological review and were evaluable for primary end point. Median age was 47 years (range, 21-74). Histologies include 17 (43%) osteosarcoma, 14 chondrosarcoma (35%, 10 conventional, four dedifferentiated [DDCS]), eight (20%) Ewing sarcoma, and one (2%) undifferentiated pleomorphic sarcoma. The PFSR at 6 months was 42% (95% confidence interval [CI], 27-58). With a median follow-up of 39.8 months (95% CI, 37.9-41.7), the median PFS and OS were 3.8 months (95% CI, 2.7-4.8) and 11.9 months (95% CI, 5.6-18.2). ORR by RECIST was 5%, with two of 38 partial responses (one of four DDCS and one of 17 osteosarcoma), 19 of 38 (50%) stable disease, and 17 of 38 (45%) progressions. Grade ≥3 adverse events were neutropenia (six of 40, 15%), anemia (5/40, hypertension (6/40, 15%), 12.5%), ALT/AST elevation (5/40, 12.5%), and pneumonitis (1/40, 2.5%). Seventeen percent of patients discontinued treatment due to toxicity, including a treatment-related grade 5 pneumonitis CONCLUSION: The trial met its primary end point in the BS cohort with >15% of patients progression-free at 6 months. However, the toxicity profile of this regimen was relevant.
Collapse
Affiliation(s)
- Emanuela Palmerini
- Osteoncology, Bone and Soft Tissue Tumors and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | | | - Andres Redondo
- Medical Oncology Department, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
| | - Nadia Hindi
- Medical Oncology Department, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
- Hospital General de Villalba, Madrid, Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Salvatore Provenzano
- Adult Mesenchymal and Rare Tumour Unit, Fondazione IRCCS Istituto Nazionale Tumori Milan, Milano, Italy
| | - Ana Sebio
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | | - Enrique de Alava
- Institute of Biomedicine of Sevilla, IBiS/Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, Seville, Spain
| | - Maria Pilar Aparisi Gomez
- Department of Radiology, Auckland City Hospital, Auckland District Health Board, Grafton, Auckland, New Zealand
- Department of Radiology, IMSKE, Valencia, Spain
| | - Lorenzo D'Ambrosio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
- Department of Oncology, University of Turin, Turin, Italy
| | - Paola Collini
- Soft Tissue Tumor Pathology, Advanced Diagnostics Department, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alberto Bazzocchi
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - David S Moura
- Medical Oncology Department, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Tumors and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal and Rare Tumour Unit, Fondazione IRCCS Istituto Nazionale Tumori Milan, Milano, Italy
| | - Javier Martin Broto
- Medical Oncology Department, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
- Hospital General de Villalba, Madrid, Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| |
Collapse
|
14
|
Mavroeidis L, Napolitano A, Huang P, Jones RL. Novel Therapeutics in Soft Tissue Sarcoma. Cancers (Basel) 2024; 17:10. [PMID: 39796641 PMCID: PMC11718850 DOI: 10.3390/cancers17010010] [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: 11/17/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
There has been noteworthy progress in molecular characterisation and therapeutics in soft tissue sarcomas. Novel agents have gained regulatory approval by the FDA. Examples are the tyrosine kinase inhibitors avapritinib and ripretinib in gastrointestinal stromal tumours (GIST), the immune check point inhibitor atezolizumab in alveolar soft part tissue sarcoma, the γ-secretase inhibitor nirogacestat in desmoid tumours, the NTRK inhibitors larotrectinib and entrectinib in tumours with NTRK fusions, the mTOR inhibitor nab-sirolimus in PEComa, and the EZH-2 inhibitor tazemetostat in epithelioid sarcoma. The FDA has also recently granted accelerated approval for autologous T-cell therapy with afami-cel in patients with HLA-A*02 and MAGE-A4-expressing synovial sarcoma. There are other promising treatments that are still investigational, such as MDM2 and CDK4/6 inhibitors in well-/dedifferentiated liposarcoma, immune checkpoint inhibitors in the head and neck angiosarcoma and a subset of patients with undifferentiated pleomorphic sarcoma, and PARP inhibitors in leiomyosarcoma. The challenges in drug development in soft tissue sarcoma are due to the rarity and the molecular heterogeneity of the disease and the fact that many subtypes are associated with complex karyotypes or non-targetable molecular alterations. We believe that progress maybe possible with a better understanding of the complex biology, the development of novel compounds for difficult targets such as proteolysis targeting chimeras (Protacs), the utilisation of modern clinical trial designs, and enhanced collaboration of academia with industry to develop treatments with a strong biologic rationale.
Collapse
Affiliation(s)
- Leonidas Mavroeidis
- Sarcoma Unit, The Royal Marsden Hospital and Institute of Cancer Research, London SW3 6JZ, UK
| | | | | | | |
Collapse
|
15
|
Movva S, Seier K, Avutu V, Banks LB, Chan J, Chi P, Dickson MA, Gounder MM, Kelly CM, Keohan ML, Maki R, Rosenbaum E, Salcito T, Rodriguez K, Dempsey R, Meyers PA, Cohen SM, Hensley ML, Konner JA, Schram AM, Lefkowitz RA, Erinjeri JP, Qin LX, Tap WD, D'Angelo SP. Histology-Specific Clinical Trial of Lenvatinib and Pembrolizumab in Patients with Sarcoma. Clin Cancer Res 2024; 30:5612-5619. [PMID: 39405335 PMCID: PMC11730159 DOI: 10.1158/1078-0432.ccr-24-2519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/20/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE Survival of patients with metastatic sarcoma remains poor, and there is a pressing need for new therapies. Most sarcoma subtypes are not responsive to immune checkpoint inhibition alone. Lenvatinib, a multireceptor tyrosine kinase inhibitor targeting tumor vasculature, has an immunomodulatory activity that contributes to its antitumor effects. Therefore, we hypothesized that a combination of lenvatinib and pembrolizumab would lead to improved clinical outcomes in patients with sarcoma. PATIENTS AND METHODS This was an open-label, single-arm study of lenvatinib and pembrolizumab in the following cohorts: (A) leiomyosarcoma, (B) undifferentiated pleomorphic sarcoma (UPS), (C) vascular sarcomas (angiosarcoma and epithelioid hemangioendothelioma), (D) synovial sarcoma or malignant peripheral nerve sheath tumor (MPNST), and (E) bone sarcomas (osteosarcoma and chondrosarcoma). The primary endpoint was the best overall response (BOR) rate documented by RECIST v1.1 by 27 weeks in each cohort, with a threshold of ≥2 responses among 10 patients. Secondary endpoints included progression-free survival, overall survival, duration of response, and safety. RESULTS Forty-six patients were evaluable for the primary endpoint, which was met in the UPS and MPNST/synovial cohorts (BOR rates by 27 weeks of 25% and 30%, respectively). There were seven partial responses overall with additional responses noted in angiosarcoma and osteosarcoma. Treatment-related adverse events of any grade and grade 3 or higher occurred in 50/51 (98%) and 29/51 (57%) of patients, respectively. CONCLUSIONS We observed durable responses in MPNST, synovial sarcoma, and osteosarcoma. Patients with UPS and angiosarcoma also responded. Further exploration of this approach is warranted to confirm activity and determine optimal dosing schedules.
Collapse
Affiliation(s)
- Sujana Movva
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Kenneth Seier
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Viswatej Avutu
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lauren B. Banks
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jason Chan
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ping Chi
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mark A. Dickson
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Mrinal M. Gounder
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ciara M. Kelly
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Mary L. Keohan
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert Maki
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Evan Rosenbaum
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Tiffany Salcito
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kaithleen Rodriguez
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rebecca Dempsey
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Paul A. Meyers
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Seth M. Cohen
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Martee L. Hensley
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jason A. Konner
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alison M. Schram
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert A. Lefkowitz
- Body Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | - Joseph P. Erinjeri
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Cornell Medical College, New York, NY
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William D. Tap
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Sandra P. D'Angelo
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
16
|
Tan Z, Wu Y, Fan Z, Gao T, Guo W, Bai C, Xue R, Li S, Zhang L, Wang X, Jia L, Liu J. Anlotinib plus TQB2450, a PD-L1 Antibody, in Patients with Advanced Alveolar Soft Part Sarcoma: A Single-Arm, Phase II Trial. Clin Cancer Res 2024; 30:5577-5583. [PMID: 39453774 DOI: 10.1158/1078-0432.ccr-24-2444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/22/2024] [Accepted: 10/23/2024] [Indexed: 10/27/2024]
Abstract
PURPOSE Alveolar soft part sarcoma (ASPS) is an ultrarare soft-tissue sarcoma with a high rate of metastasis and no established treatment. This study aimed to explore the efficacy and safety of anlotinib (a tyrosine kinase inhibitor) and TQB2450 (a PD-L1 inhibitor) in patients with ASPS. PATIENTS AND METHODS This single-arm, phase II study evaluated the efficacy of TQB2450, an anti-PD-L1 agent, combined with anlotinib, a tyrosine kinase inhibitor, in adults with advanced ASPS. TQB2450 was given intravenously (1,200 mg) on day 1, and anlotinib (12 mg/day) was taken orally from day 1 to 14 every 3 weeks. The primary endpoint was overall response rate, with secondary endpoints including duration of response, progression-free survival, and overall survival. Lymphocyte infiltration and tertiary lymphoid structure (TLS) were also analyzed as potential prognostic biomarkers. RESULTS The study enrolled 29 patients, of whom 28 were evaluable (one withdrew because of acute pancreatitis). An objective response was achieved in 82.1% of patients, including 4 complete and 19 partial responses. The median time to response was 2.8 months, and the duration of response was not reached, with an estimated median progression-free survival of 35.2 months. Grade 3 to 4 treatment-related adverse events occurred in 44.8% of patients, with no study-related deaths. Responders had a higher proportion of TLS area, TLS density, and CD20-positive immune cells. CONCLUSIONS The combination of anlotinib and TQB2450 is effective and tolerable in patients with ASPS. TLS may serve as a prognostic biomarker, meriting further investigation.
Collapse
Affiliation(s)
- Zhichao Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhengfu Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Tian Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| | - Chujie Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ruifeng Xue
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lu Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinyu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ling Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
17
|
Fernandes S, Rastogi S, Bhatia KP, Chitikela S, Shamim SA, Gammanagatti S, Barwad A. Real world outcomes in alveolar soft part sarcomas: experience with an ultra-rare sarcoma from a tertiary care centre in North India. Ecancermedicalscience 2024; 18:1813. [PMID: 40171452 PMCID: PMC11959123 DOI: 10.3332/ecancer.2024.1813] [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: 07/08/2024] [Indexed: 04/03/2025] Open
Abstract
Background Alveolar soft part sarcoma (ASPS) is a rare, indolent soft tissue sarcoma, with a high predilection for systemic dissemination. This study aims to elucidate the patterns of clinical presentation of ASPS and their treatment outcomes, with emphasis on the use of newer therapeutic agents and their efficacy in advanced ASPS. Methods This was a retrospective cohort that included patients with ASPS treated at our institute between 2016 and 2023. Clinicopathological data were obtained from case records and analysed to assess outcomes. Results The study included 34 patients (19 males, 15 females) with a median age of 28 (3-72) years. 7 patients presented with localised disease, and 27 with metastatic disease. The most common site of primary was the extremities (73%), and the most common sites of metastasis included the lungs (82%) and bones (21%). Brain metastasis was seen in 7 patients at baseline (25.9%). 90% of patients with metastatic disease received a tyrosine kinase inhibitor in the first-line setting with a median progression-free survival of 12 months. The median overall survival in this subset was 36 months. 7 patients with the advanced disease received immune-checkpoint inhibitors (ICIs) (3-atezolizumab, 4-nivolumab); 2 patients on atezolizumab and 1 on nivolumab continue to be progression free at 20,15 and 52 months, respectively. Patients with brain metastasis were seen to have markedly poor outcomes. Conclusion The use of anti-angiogenic agents and ICIs has significantly improved survival in patients with advanced ASPS. Brain metastasis confers poor survival in these patients despite the use of newer agents. This study represents the largest cohort of patients with advanced ASPS from this region.
Collapse
Affiliation(s)
- Sanal Fernandes
- Department of Medical Oncology, AIIMS, New Delhi 110029, India
| | - Sameer Rastogi
- Department of Medical Oncology, AIIMS, New Delhi 110029, India
| | | | | | - Shamim A Shamim
- Department of Nuclear Medicine, AIIMS, New Delhi 110029, India
| | | | - Adarsh Barwad
- Department of Pathology, AIIMS, New Delhi 110029, India
| |
Collapse
|
18
|
Moura DS, Lopez-Marti JM, Benesova I, de Andrea C, di Lernia D, Lacerenza S, Mondaza-Hernandez JL, Martin-Ruiz M, Ramirez-Calvo M, Grignani G, Martinez-Trufero J, Redondo A, Valverde C, Stacchiotti S, Lopez-Pousa A, Lopez-Guerrero JA, Gutierrez A, Encinas-Tobajas V, Hindi N, Sangiolo D, Lopez-Martin JA, Strizova ZO, Martin-Broto J. Predictive and Dynamic Signature for Antiangiogenics in Combination with a PD1 Inhibitor in Soft-Tissue Sarcoma: Correlative Studies Linked to the IMMUNOSARC Trial. Clin Cancer Res 2024; 30:5192-5206. [PMID: 39283727 DOI: 10.1158/1078-0432.ccr-24-1782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE The IMMUNOSARC trial combined an antiangiogenic agent (sunitinib) with a PD1 inhibitor (nivolumab) in advanced sarcomas. Here, we present the first correlative studies of the soft-tissue sarcoma cohort enrolled in this trial. EXPERIMENTAL DESIGN Formalin-fixed paraffin-embedded and peripheral blood samples were collected at baseline and week 13. Formalin-fixed paraffin-embedded samples were used for transcriptomics and multiplex immunofluorescence, whereas peripheral blood samples were used for multiplexed immunoassays. Flow cytometry and Luminex assays were performed to validate translational findings in tumor-isolated cells and peripheral blood mononuclear cells derived from patients. RESULTS The density of intratumoral CD8+ T cells, measured by multiplexed immunophenotyping, was significantly increased after treatment. This augment was accompanied by the dynamic significant increase in the gene expressions of CD86, CHI3L1, CXCL10, CXCL9, LAG3, and VCAM1 and the decrease in the expression levels of NR4A1. In peripheral blood, 12 proteins were significantly modulated by treatment at week 13. A score integrating the dynamic expression of the 7 genes and the 12 soluble factors separated 2 groups with distinct progression-free survival (PFS): 4.1 months [95% confidence interval, 3.5-not reached (NR)] versus 17 months (95% confidence interval, 12.0-NR), P = 0.014. This molecular score was predictive of PFS when applied to the normalized data determined in the baseline samples. CONCLUSIONS Treatment with sunitinib and nivolumab inflamed the sarcoma microenvironment, increasing CD8+ T-cell density and the expression of several genes/proteins with relevance in the response to PD1 inhibitors. A molecular signature identified two groups of patients with distinct PFS for the combination of antiangiogenics plus PD1 inhibitor therapy.
Collapse
Affiliation(s)
- David S Moura
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Jesus M Lopez-Marti
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Carlos de Andrea
- Department of Pathology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Davide di Lernia
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Serena Lacerenza
- Institute of Biomedicine of Seville (IBiS; US, CSIC, HUVR), Seville, Spain
| | | | - Marta Martin-Ruiz
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | | | - Giovanni Grignani
- Medical Oncology Unit, Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Andres Redondo
- Department of Medical Oncology, University Hospital La Paz, Madrid, Spain
| | - Claudia Valverde
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - José A Lopez-Guerrero
- Fundación Institute Valenciano of Oncology, Valencia, Spain
- Joint Cancer Research Unit IVO-CIPF, Valencia, Spain
- Department of Pathology, Medical School of the Catholic University of Valencia, Valencia, Spain
| | - Antonio Gutierrez
- Department of Hematology, University Hospital Son Espases, Mallorca, Spain
| | | | - Nadia Hindi
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
- Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy
| | - Jose A Lopez-Martin
- Department of Medical Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Zuzana Ozaniak Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Javier Martin-Broto
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
- Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| |
Collapse
|
19
|
Mondaza-Hernandez JL, Hindi N, Fernandez-Serra A, Ramos R, Gonzalez-Cámpora R, Gómez-Mateo MC, Martinez-Trufero J, Lavernia J, Lopez-Pousa A, Laínez N, Martinez-Garcia J, Valverde C, Vaz-Salgado MÁ, Garcia-Plaza G, Marin-Borrero I, Carrillo-Garcia J, Martin-Ruiz M, Romero P, Gutierrez A, López-Guerrero JA, Moura DS, Martin-Broto J. Exploratory analysis of immunomodulatory factors identifies L1CAM as a prognostic marker in alveolar soft-part sarcoma. Ther Adv Med Oncol 2024; 16:17588359241293951. [PMID: 39502403 PMCID: PMC11536517 DOI: 10.1177/17588359241293951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Background Alveolar soft-part sarcoma (ASPS) is a rare tumor driven by the ASPSCR1-TFE3 fusion protein, with a propensity for metastasis. Prognostic factors remain poorly understood, and traditional chemotherapies are largely ineffective. Recent interest lies in immune checkpoint inhibitors (ICIs), yet predictive biomarkers for treatment response are lacking. Previous studies have shown promising results with ICIs in ASPS, indicating a need for further investigation into biomarkers associated with immune response. Objectives To identify prognostic biomarkers in ASPS and to explore the role of immune-related markers, particularly L1CAM, in predicting patient outcomes. Design A retrospective cohort study of 19 ASPS patients registered in the GEIS database. The study involved the collection of clinical and histopathological data, followed by an analysis of immune markers and gene expression profiles to identify potential prognostic indicators. Methods Clinical and histopathological data were retrospectively collected from the GEIS-26 study cohort of 19 ASPS patients. Immunohistochemistry was performed to evaluate immune markers programmed death-1 ligand (PD-L1), programmed death-1, FAS, FASL, CD8, CD3, and CD4. An HTG ImmunOncology panel was conducted on formalin-fixed paraffin-embedded samples to explore gene expression. Effects of differentially expressed genes on survival were explored by Kaplan-Meier. Results PD-L1 positivity was widely observed (63%) in tumors, and CD8+ lymphocytic infiltration was common. High CD8 density correlated with greater overall survival (OS) while not statistically significant. No associations were found for other immune markers. L1CAM was identified as differentially expressed in patients with low CD8 infiltration and correlated negatively with OS. Conclusion High L1CAM expression correlated with poorer OS, highlighting its potential as a prognostic marker and therapeutic target in ASPS. Immunomodulatory interventions may hold promise, as evidenced by PD-L1 expression and CD8+ infiltration. Further research, including larger cohorts and international collaborations, is needed to validate these findings and explore therapeutic strategies targeting L1CAM in ASPS.
Collapse
Affiliation(s)
- José L. Mondaza-Hernandez
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Nadia Hindi
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | | | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Mallorca, Spain
| | | | | | | | - Javier Lavernia
- Medical Oncology Department, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | | | - Nuria Laínez
- Department of Medical Oncology, Complejo Hospitalario de Navarra Pamplona, Spain
| | | | - Claudia Valverde
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | - María Ángeles Vaz-Salgado
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, CIBERONC, Madrid, Spain
| | - Gabriel Garcia-Plaza
- Department of Surgery, Hospital Universitario Insular, Las Palmas de Gran Canaria, Spain
| | - Isabel Marin-Borrero
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Jaime Carrillo-Garcia
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Marta Martin-Ruiz
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Pablo Romero
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Antonio Gutierrez
- Hematology Department, Hospital Son Espases/IdISBa, Palma de Mallorca, Spain
| | - Jose A. López-Guerrero
- Molecular Biology Department, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | - David S. Moura
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Javier Martin-Broto
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Reyes Católicos 2, Madrid 28040, Spain
- University Hospital General de Villalba, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| |
Collapse
|
20
|
Ng VY, Sahlani MN, Fogel JD, Chiu AK, Kallen ME, Davis D, Snider J, Regine W, Bentzen SM, Sausville E. Results of an Integrated Phase I/II Prospective Clinical Trial (NEXIS) for Neoadjuvant Anti-PD-L1 (Durvalumab) and Anti-CTLA-4 (Tremelimumab) With Radiation for High-Risk Soft-Tissue Sarcoma of the Trunk and Extremities. Cureus 2024; 16:e72119. [PMID: 39575028 PMCID: PMC11581454 DOI: 10.7759/cureus.72119] [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] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Background The current management of large, high-grade soft tissue sarcoma (STS) of the trunk and extremities includes radiation and surgical resection. The initial use of chemotherapy and targeted therapy are controversial and although most patients present with localized disease, many eventually develop incurable metastases. The results and analysis of the safety and antitumor activity of combined checkpoint inhibitor immunotherapy with neoadjuvant radiation for high-risk primary STS are presented here. Methods This was an integrated phase I/II prospective single-arm trial (Nutrition and Exercise in Critical Illness Trial (NEXIS) trial). Eligible patients were age ≥18 years with histologically confirmed intermediate or high-grade STS of the trunk or extremity ≥5 cm diameter and were Eastern Cooperative Oncology Group performance status 0-1. The treatment algorithm included neoadjuvant anti-PD-L1 (Durvalumab) and anti-CTLA-4 (Tremelimumab) for three cycles of four weeks/cycle along with external beam radiation for five weeks, followed by wide surgical resection, and adjuvant Durvalumab monotherapy for four cycles. High-grade toxicity was continually assessed for the first 12 patients in phase I and the primary endpoint for phase II was an excellent histological response (grade 0 or 1 score on a semi-quantitative assessment for tumor regression). This study was registered with ClinicalTrials.gov, number NCT03116529. Findings Between October 2017 and November 2021, 23 patients were enrolled. Five patients had progression of distant disease during neoadjuvant treatment and withdrew from the study before surgery. A total of 18 patients who completed at least the neoadjuvant immunotherapy, radiation and surgery were included for analysis. The most common tumor was undifferentiated pleomorphic sarcoma (n=9, 50%). The occurrence of any adverse event (AE) was recorded in 16 (88.9%) patients, and 3 (16.7%) patients had a serious AE. Eight out of 18 patients (44.4%) had disease-free survival at a median of 39.7 months. Four out of 18 patients (22.2%) were alive-with-disease at a median of 37.1 months from diagnosis of distant metastasis, and six out of 18 (33.3%) died of disease at a median of 20.8 months from diagnosis of distant metastasis. Local recurrence occurred in two patients (11.1%) and was concomitant with distant disease in each case. Based on Response Evaluation Criteria in Solid Tumors v1.1, a partial response was noted in five (27.8%) cases, stable disease in 10 (55.6%) cases, and progressive disease in three (16.7%) cases. Histological semiquantitative analysis revealed a "good" response in eight (44.4%) patients, a "moderate" response in four (22.2%) patients, and a "poor" response in six (33.3%) patients. The mean patient-reported outcome measures regarding fatigue, physical function, or physical interference demonstrated no significant differences between various timepoints before, during, or after treatment. Conclusion Neoadjuvant combined immunotherapy and radiation for high-risk STS was relatively well-tolerated. The histological, radiologic, and clinical outcome data in this novel trial were relatively similar to historical literature for non-immunotherapy treatment regimens.
Collapse
Affiliation(s)
- Vincent Y Ng
- Department of Orthopedics, University of Maryland Medical Center, Baltimore, USA
| | - Mario N Sahlani
- Department of Orthopedics, University of Maryland Medical Center, Baltimore, USA
| | - Jessa D Fogel
- Department of Orthopedics, University of Maryland Medical Center, Baltimore, USA
| | - Anthony K Chiu
- Department of Orthopedics, University of Maryland Medical Center, Baltimore, USA
| | - Michael E Kallen
- Department of Pathology, University of Maryland School of Medicine, Baltimore, USA
| | - Derik Davis
- Department of Diagnostic Radiology, University of Maryland Medical Center, Baltimore, USA
| | - James Snider
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | - William Regine
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | - Søren M Bentzen
- Greenebaum Comprehensive Cancer Center and Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, USA
| | - Edward Sausville
- Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
21
|
Yu S, Yao X. Advances on immunotherapy for osteosarcoma. Mol Cancer 2024; 23:192. [PMID: 39245737 PMCID: PMC11382402 DOI: 10.1186/s12943-024-02105-9] [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: 07/20/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024] Open
Abstract
Osteosarcoma is the most common primary bone cancer in children and young adults. Limited progress has been made in improving the survival outcomes in patients with osteosarcoma over the past four decades. Especially in metastatic or recurrent osteosarcoma, the survival rate is extremely unsatisfactory. The treatment of osteosarcoma urgently needs breakthroughs. In recent years, immunotherapy has achieved good therapeutic effects in various solid tumors. Due to the low immunogenicity and immunosuppressive microenvironment of osteosarcoma, immunotherapy has not yet been approved in osteosarcoma patients. However, immune-based therapies, including immune checkpoint inhibitors, chimeric antigen receptor T cells, and bispecfic antibodies are in active clinical development. In addition, other immunotherapy strategies including modified-NK cells/macrophages, DC vaccines, and cytokines are still in the early stages of research, but they will be hot topics for future study. In this review, we showed the functions of cell components including tumor-promoting and tumor-suppressing cells in the tumor microenvironment of osteosarcoma, and summarized the preclinical and clinical research results of various immunotherapy strategies in osteosarcoma, hoping to provide new ideas for future research in this field.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xudong Yao
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
22
|
Patel AJ, Hemead H, Jesani H, Bille A, Taniere P, Middleton G. Squamous Cell Carcinoma in Never Smokers: An Insight into SMARCB1 Loss. Int J Mol Sci 2024; 25:8165. [PMID: 39125735 PMCID: PMC11311737 DOI: 10.3390/ijms25158165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Lung cancer remains the leading cause of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) constituting 85% of cases. Among NSCLCs, squamous cell carcinoma (SqCC) is strongly associated with smoking. However, lung cancer in never smokers (LCINS) represents approximately 25% of lung cancer cases globally and shows increasing incidence, particularly in East Asia. LCINS-SqCC is less well-characterized, especially regarding its genomic alterations and their impact on clinical outcomes. We conducted a retrospective analysis over a 20-year period (July 2003-July 2023) at two major tertiary centers in the UK. The cohort included 59 patients with LCINS-SqCC who underwent radical surgical resection. Data collected included demographic information, comorbidities, histopathological details, and outcome metrics such as disease-free and overall survival. Molecular sequencing of tumor specimens was performed to identify genomic aberrations. The cohort had a median age of 71 years (IQR 62-77) and a median BMI of 25.4 (IQR 22.8-27.8), with a slight male predominance (53%). The majority of patients (93%) had a preoperative MRC of 1-2. Recurrent disease was observed in 23 patients (39%), and 32 patients (54%) had died at a median follow-up of 3 years. Median disease-free survival was 545 days (IQR 132-1496), and overall survival was 888 days (IQR 443-2071). Preoperative creatinine levels were higher in patients who experienced recurrence (p = 0.037). Molecular analysis identified biallelic SMARCB1 loss in two younger patients, associated with rapid disease progression despite R0 resection. These patients' tumors were PDL1-negative, TTF-1-negative, and positive for cytokeratin, CD56, and p40. SMARCB1-deficient SqCC in never smokers represents a highly aggressive variant with poor disease-free survival, highlighting the importance of integrating advanced molecular diagnostics in clinical practice. This study underscores the necessity for personalized treatment strategies, including targeted therapies such as EZH2 inhibitors and immune checkpoint blockade, to address the unique molecular pathways in SMARCB1-deficient cancers. Further clinical trials are essential to optimize therapeutic approaches for this challenging subgroup of lung cancer.
Collapse
Affiliation(s)
- Akshay J. Patel
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK;
- Department of Thoracic Surgery, University Hospitals Birmingham, Birmingham B15 2GW, UK; (H.H.); (H.J.)
| | - Hanan Hemead
- Department of Thoracic Surgery, University Hospitals Birmingham, Birmingham B15 2GW, UK; (H.H.); (H.J.)
| | - Hannah Jesani
- Department of Thoracic Surgery, University Hospitals Birmingham, Birmingham B15 2GW, UK; (H.H.); (H.J.)
| | - Andrea Bille
- Department of Thoracic Surgery, Guy’s Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London SE1 9RT, UK;
| | - Philippe Taniere
- Department of Cellular Histopathology, University Hospitals Birmingham, Birmingham B15 2GW, UK;
| | - Gary Middleton
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK;
| |
Collapse
|
23
|
Grice S, Olsson-Brown A, Naisbitt DJ, Hammond S. Immunological Drug-Drug Interactions Affect the Efficacy and Safety of Immune Checkpoint Inhibitor Therapies. Chem Res Toxicol 2024; 37:1086-1103. [PMID: 38912648 PMCID: PMC11256900 DOI: 10.1021/acs.chemrestox.4c00067] [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: 02/20/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024]
Abstract
With the rapid expansion in the development and clinical utility of immune checkpoint inhibitors (ICIs) for oncology, the continual evaluation of the safety profile of such agents is imperative. The safety profile of ICIs as monotherapy is dominated by immune-related adverse events, which can be considered as an extension of the mechanism of action of these immunomodulatory drugs. Further to this, an emerging theme is that ICI treatment can significantly impact upon the tolerability of coadministered medications. Numerous reports in literature indicate that ICIs may alter the immunological perception of coadministered drugs, resulting in undesirable reactions to a variety of concomitant medications. These reactions can be severe in manifestation, including hepatotoxicity and Stevens-Johnson Syndrome (SJS)/toxic epidermal necrolysis (TEN), but may also have detrimental impact on malignancy control. To minimize the impact of such drug-drug interactions on patients, it is imperative to identify medications that may cause these reactions, understand the underlying mechanisms, consider the timing and dosing of comedication, and explore alternative medications with comparable efficacies. Improving our understanding of how concomitant medications affect the safety and efficacy of ICIs can allow for potential culprit drugs to be identified/removed/desensitized. This approach will allow the continuation of ICI therapy that may have been discontinued otherwise, thereby improving malignant control and patient and drug development outcomes.
Collapse
Affiliation(s)
- Sophie Grice
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Anna Olsson-Brown
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
- Sussex
Cancer Centre, University Hospitals Sussex, Brighton BN2 5BD, U.K.
| | - Dean J. Naisbitt
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Sean Hammond
- Department
of Molecular and Clinical Pharmacology, Institute of Translational
Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
- ApconiX, Alderley Edge SK10 4TG, U.K.
| |
Collapse
|
24
|
Sicinska E, Sudhakara Rao Kola V, Kerfoot JA, Taddei ML, Al-Ibraheemi A, Hsieh YH, Church AJ, Landesman-Bollag E, Landesman Y, Hemming ML. ASPSCR1::TFE3 Drives Alveolar Soft Part Sarcoma by Inducing Targetable Transcriptional Programs. Cancer Res 2024; 84:2247-2264. [PMID: 38657118 PMCID: PMC11250573 DOI: 10.1158/0008-5472.can-23-2115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 02/09/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Alveolar soft part sarcoma (ASPS) is a rare mesenchymal malignancy driven by the ASPSCR1::TFE3 fusion. A better understanding of the mechanisms by which this oncogenic transcriptional regulator drives cancer growth is needed to help identify potential therapeutic targets. In this study, we characterized the transcriptional and chromatin landscapes of ASPS tumors and preclinical models, identifying the essential role of ASPSCR1::TFE3 in tumor cell viability by regulating core transcriptional programs involved in cell proliferation, angiogenesis, and mitochondrial biology. ASPSCR1::TFE3 directly interacted with key epigenetic regulators at enhancers and promoters to support ASPS-associated transcription. Among the effector programs driven by ASPSCR1::TFE3, cell proliferation was driven by high levels of cyclin D1 expression. Disruption of cyclin D1/CDK4 signaling led to a loss of ASPS proliferative capacity, and combined inhibition of CDK4/6 and angiogenesis halted tumor growth in xenografts. These results define the ASPS oncogenic program, reveal mechanisms by which ASPSCR1::TFE3 controls tumor biology, and identify a strategy for therapeutically targeting tumor cell-intrinsic vulnerabilities. Significance: The ASPSCR1::TFE3 fusion propels the growth of alveolar soft part sarcoma by activating transcriptional programs that regulate proliferation, angiogenesis, mitochondrial biogenesis, and differentiation and can be therapeutically targeted to improve treatment.
Collapse
MESH Headings
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Sarcoma, Alveolar Soft Part/genetics
- Sarcoma, Alveolar Soft Part/pathology
- Sarcoma, Alveolar Soft Part/metabolism
- Humans
- Animals
- Mice
- Cell Proliferation/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Gene Expression Regulation, Neoplastic
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cyclin-Dependent Kinase 4/genetics
- Cyclin-Dependent Kinase 4/metabolism
- Cyclin-Dependent Kinase 4/antagonists & inhibitors
- Female
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Ewa Sicinska
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Vijaya Sudhakara Rao Kola
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Joseph A. Kerfoot
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Madeleine L. Taddei
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Alyaa Al-Ibraheemi
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi-Hsuan Hsieh
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Alanna J. Church
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Esther Landesman-Bollag
- Department of Medicine, Section of Hematology and Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Yosef Landesman
- Cure Alveolar Soft Part Sarcoma International, Brookline, Massachusetts, USA
| | - Matthew L. Hemming
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
25
|
Czarnecka AM, Chmiel P, Błoński PJ, Świtaj T, Rogala P, Falkowski S, Koseła-Paterczyk H, Teterycz P, Morysiński T, Spałek M, Wągrodzki M, Rutkowski P. Real-world outcomes of metastatic clear cell sarcoma sequential chemotherapy. J Chemother 2024:1-12. [PMID: 38973299 DOI: 10.1080/1120009x.2024.2372524] [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: 04/18/2024] [Accepted: 06/21/2024] [Indexed: 07/09/2024]
Abstract
Clear cell sarcoma is an ultra-rare chemoresistant subtype of soft tissue sarcoma. This retrospective analysis aimed to clarify the efficacy of palliative chemotherapy in CCS by assessing response rates, progression-free survival (PFS), and overall survival (OS) at a referral center. A retrospective analysis of palliative treatment was conducted on patients with CCS treated at the sarcoma unit from 1997 to 2023. Treatment responses were assessed using RECIST criteria, and the Kaplan-Meier method was used to calculate PFS and OS. The analysis covered 23 CCS chemotherapy-treated patients with 11 (47.8%) men. The median age at the palliative treatment start was 32 years (range 18-59). The median follow-up was 8.2 months. Four patients were referred to our centre for M1 disease, and 6 received perioperative chemotherapy and progressed during follow-up. In the first line, 14 patients received anthracycline-based chemotherapy (60.9%), five were treated with ifosfamide (HD-IFO), and four received other regimens. One patient (4.3%) achieved partial response (PR), and 12 patients (52.2%) achieved stable disease (SD) as the best response. Median PFS in 1 line was 2.79 months (95% CI: 2.04-8.38), and 1.76 months (95% CI: 0.72-6.97) in the second line. The median OS from first-line palliative chemotherapy was 8.2 months (95% CI: 6.2-14), and the second-line palliative chemotherapy mOS was 4.6 months (95% CI: 3.9-NA). Perioperatively anthracycline-pretreated worsened patients' median PFS in the M1 setting. Poor responses to conventional chemotherapy were observed in CCS, indicating a need for further clinical trials in this indication.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Paulina Chmiel
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Medical Faculty, Medical University of Warsaw, Warsaw, Poland
| | - Piotr J Błoński
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Medical Faculty, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Świtaj
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paweł Rogala
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Sławomir Falkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Hanna Koseła-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paweł Teterycz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Computational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Tadeusz Morysiński
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Mateusz Spałek
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Radiotherapy I, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michał Wągrodzki
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
26
|
Martin-Broto J, Hindi N, Moura DS. Combination treatment with PD1/PDL-1 inhibitors for sarcomas: state of the art, next questions. Curr Opin Oncol 2024; 36:269-275. [PMID: 38726845 DOI: 10.1097/cco.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW Only a small fraction of sarcomas exhibit recognized parameters of immune sensitivity, such as tumor mutational burden, PDL-1 expression, or microsatellite instability. Combined strategies aimed to modulate tumor microenvironment to increase the efficacy of PD1/PDL-1 inhibitors in sarcoma. Most explored prospective studies were based on combinations of PD1/PDL-1 inhibitors with antiangiogenics, other immune checkpoints, or chemotherapy. RECENT FINDINGS Results on 6-month PFS rate, median PFS, and ORR in trials using PD1/PDL-1 inhibitors plus antiangiogenics ranged respectively as 46.9-55%, 4.7-7.8 months and 21-36.7%. In combination with other immune checkpoint inhibitors, the results of median PFS and ORR ranged from 2.8-4.1 months and 10-16%, respectively. In combination with chemotherapy, the best results were obtained with doxorubicin-based regimens compared to other agents. Duplet-based chemotherapy plus anti-PD1/PDL-1 obtained the highest ORR (56.2%) compared with doxorubicin (19-36.7%). Currently, the most robust predictive biomarker for anti-PD1/PDL-1 efficacy is the presence of tertiary lymphoid structures (TLS) with mature dendritic cells. SUMMARY Even when direct comparisons between PD1/PDL-1 inhibitor-based combinations and single agents have not been performed yet in sarcoma, some combinations appear promising. Studies controlling heterogeneity by biomarker or histotype selection contribute to an increase in efficacy or knowledge crucial for future comparative trials.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital
- University Hospital General de Villalba
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Nadia Hindi
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital
- University Hospital General de Villalba
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - David S Moura
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| |
Collapse
|
27
|
Ingangi V, De Chiara A, Ferrara G, Gallo M, Catapano A, Fazioli F, Di Carluccio G, Peranzoni E, Marigo I, Carriero MV, Minopoli M. Emerging Treatments Targeting the Tumor Microenvironment for Advanced Chondrosarcoma. Cells 2024; 13:977. [PMID: 38891109 PMCID: PMC11171855 DOI: 10.3390/cells13110977] [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: 04/30/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Chondrosarcoma (ChS), a malignant cartilage-producing tumor, is the second most frequently diagnosed osseous sarcoma after osteosarcoma. It represents a very heterogeneous group of malignant chemo- and radiation-resistant neoplasms, accounting for approximately 20% of all bone sarcomas. The majority of ChS patients have a good prognosis after a complete surgical resection, as these tumors grow slowly and rarely metastasize. Conversely, patients with inoperable disease, due to the tumor location, size, or metastases, represent a great clinical challenge. Despite several genetic and epigenetic alterations that have been described in distinct ChS subtypes, very few therapeutic options are currently available for ChS patients. Therefore, new prognostic factors for tumor progression as well as new treatment options have to be explored, especially for patients with unresectable or metastatic disease. Recent studies have shown that a correlation between immune infiltrate composition, tumor aggressiveness, and survival does exist in ChS patients. In addition, the intra-tumor microvessel density has been proven to be associated with aggressive clinical behavior and a high metastatic potential in ChS. This review will provide an insight into the ChS microenvironment, since immunotherapy and antiangiogenic agents are emerging as interesting therapeutic options for ChS patients.
Collapse
Affiliation(s)
- Vincenzo Ingangi
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (V.I.); (G.D.C.); (M.M.)
| | - Annarosaria De Chiara
- Histopathology Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (A.D.C.); (G.F.)
| | - Gerardo Ferrara
- Histopathology Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (A.D.C.); (G.F.)
| | - Michele Gallo
- Musculoskeletal Surgery Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (M.G.); (A.C.); (F.F.)
| | - Antonio Catapano
- Musculoskeletal Surgery Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (M.G.); (A.C.); (F.F.)
| | - Flavio Fazioli
- Musculoskeletal Surgery Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (M.G.); (A.C.); (F.F.)
| | - Gioconda Di Carluccio
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (V.I.); (G.D.C.); (M.M.)
| | - Elisa Peranzoni
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (E.P.); (I.M.)
| | - Ilaria Marigo
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (E.P.); (I.M.)
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy
| | - Maria Vincenza Carriero
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (V.I.); (G.D.C.); (M.M.)
| | - Michele Minopoli
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy; (V.I.); (G.D.C.); (M.M.)
| |
Collapse
|
28
|
Wood GE, Meyer C, Petitprez F, D'Angelo SP. Immunotherapy in Sarcoma: Current Data and Promising Strategies. Am Soc Clin Oncol Educ Book 2024; 44:e432234. [PMID: 38781557 DOI: 10.1200/edbk_432234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Traditionally sarcomas have been considered immunologically quiet tumours, with low tumour mutational burden (TMB) and an immunosuppressive tumour microenvironment (TME), consisting of decreased T-cell infiltration and elevated levels of H1F1α, macrophages and neutrophils.1,2 However, research has shown that a subset of sarcomas are immunologically 'hot' with either high TMB, PDL-1 expression, CD8+ T cells or presence of tertiary lymphoid structures (TLS) demonstrating sensitivity to immunotherapy.3,4 Here, we review the current evidence for immunotherapy use in bone sarcomas (BS) and soft tissue sarcomas (STS), with immune checkpoint inhibitors (ICI) and adoptive cellular therapies including engineered T-cell therapies, chimeric antigen receptor (CAR) T-cell therapies, tumour infiltrating lymphocytes (TILs) and cancer vaccines and biomarkers of response.
Collapse
Affiliation(s)
- Georgina E Wood
- University College Hospital of London, London, United Kingdom
| | | | | | | |
Collapse
|
29
|
Honkisz-Orzechowska E, Barczyk-Woźnicka O, Kaleta M, Handzlik J, Kieć-Kononowicz K. Studies on Autophagy and Apoptosis of Fibrosarcoma HT-1080 Cells Mediated by Chalcone with Indole Moiety. Int J Mol Sci 2024; 25:6100. [PMID: 38892288 PMCID: PMC11172467 DOI: 10.3390/ijms25116100] [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: 04/15/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
This study demonstrated the anticancer efficacy of chalcones with indole moiety (MIPP, MOMIPP) in fibrosarcoma cells for the first time. The results showed that MIPP and MOMIPP reduced the viability of HT-1080 cells in a concentration-dependent manner. MOMIPP was more active than MIPP in HT-1080 cells, showing lower IC50 values (3.67 vs. 29.90 μM). Both compounds at a concentration of 1 μM induced apoptosis in HT-1080 cells, causing death strictly related to caspase activation, as cell viability was restored when the caspase inhibitor Z-VAD was added. Reactive oxygen species production was approximately 3-fold higher than in control cells, and cotreatment with the inhibitor of mitochondrial ATPase oligomycin diminished this effect. Such effects were also reflected in mitochondrial dysfunction, including decreased membrane potential. Interestingly, the compounds that were studied caused massive vacuolization in HT-1080 cells. Immunocytochemical staining and TEM analysis showed that HT-1080 cells exhibited increased expression of the LC3-II protein and the presence of autophagosomes with a double membrane, respectively. Both compounds induced apoptosis, highlighting a promising link between autophagy and apoptosis. This connection could be a new target for therapeutic strategies to overcome chemoresistance, which is a significant cause of treatment failure and tumour recurrence in fibrosarcoma following traditional chemotherapy.
Collapse
Affiliation(s)
- Ewelina Honkisz-Orzechowska
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Kraków, Poland; (E.H.-O.); (M.K.); (J.H.)
| | - Olga Barczyk-Woźnicka
- Laboratory of Transmission Electron Microscopy, Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, 9 Gronostajowa Street, 30-387 Kraków, Poland;
| | - Maria Kaleta
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Kraków, Poland; (E.H.-O.); (M.K.); (J.H.)
| | - Jadwiga Handzlik
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Kraków, Poland; (E.H.-O.); (M.K.); (J.H.)
| | - Katarzyna Kieć-Kononowicz
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Kraków, Poland; (E.H.-O.); (M.K.); (J.H.)
| |
Collapse
|
30
|
Stacchiotti S, Bouche G, Herold R, Pantziarka P, Schuster K, Wilson R, Pignatti F, Kasper B. How to develop new systemic treatments in ultra-rare cancers with high unmet needs? The case of alveolar soft-part sarcoma. Eur J Cancer 2024; 202:114003. [PMID: 38479120 DOI: 10.1016/j.ejca.2024.114003] [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: 12/07/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/21/2024]
Abstract
Developing new drugs or generating evidence for existing drugs in new indications for ultra-rare cancers is complex and carries a high-risk of failure. This gets even harder in ultra-rare tumours, which have an annual incidence of 1 per 1,000,000 population or less. Here, we illustrate the problem of adequate evidence generation in ultra-rare tumours, using Alveolar Soft-Part Sarcomas (ASPS) - an ultra-rare sarcoma newly diagnosed in approximately 60 persons a year in the European Union - as an exemplar case showing challenges in development despite being potentially relevant for classes of agents. We discuss some possible approaches for addressing such challenges, especially focussing on constructive collaboration between academic groups, patients and advocates, drug manufacturers, and regulators to optimise drug development in ultra-rare cancers. This article, written by various European stakeholders, proposes a way forward to ultimately get better options for patients with ultra-rare cancers.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | | | | | | | | | - Roger Wilson
- Sarcoma Patient Advocacy Global Network, Wölfersheim, Germany
| | | | - Bernd Kasper
- Sarcoma Unit, Mannheim Cancer Center (MCC), Mannheim University Medical Center, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
31
|
Kyriazoglou A, Pagkali A, Kotsantis I, Economopoulou P, Kyrkasiadou M, Moutafi M, Gavrielatou N, Anastasiou M, Boulouta A, Pantazopoulos A, Giannakakou M, Digklia A, Psyrri A. Well-differentiated liposarcomas and dedifferentiated liposarcomas: Systemic treatment options for two sibling neoplasms. Cancer Treat Rev 2024; 125:102716. [PMID: 38492514 DOI: 10.1016/j.ctrv.2024.102716] [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: 10/17/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Well-differentiated liposarcomas (WDLPS) and dedifferentiated liposarcomas (DDLPS) account for 60 % of all liposarcomas, reflecting the heterogeneity of this type of sarcoma. Genetically, both types of liposarcomas are characterized by the amplification of MDM2 and CDK4 genes, which indicates an important molecular event with diagnostic and therapeutic relevance. In both localized WDLPS and DDLPS of the retroperitoneum and the extremities, between 25 % and 30 % of patients have local or distant recurrence, even when perioperatively treated, with clear margins present. The systemic treatment of WDLPS and DDLPS remains a challenge, with anthracyclines as the gold standard for first-line treatment. Several regimens have been tested with modest results regarding their efficacy. Herein we discuss the systemic treatment options for WDLPS and DDLPS and review their reported clinical efficacy results.
Collapse
Affiliation(s)
- A Kyriazoglou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece.
| | - A Pagkali
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - I Kotsantis
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - P Economopoulou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Kyrkasiadou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Moutafi
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - N Gavrielatou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Anastasiou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - A Boulouta
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - A Pantazopoulos
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Giannakakou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - A Digklia
- Sarcoma Center, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne University Lausanne, Switzerland
| | - A Psyrri
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| |
Collapse
|
32
|
Liao Z, Teng J, Li T, Liu H, Li T, Zhang C, Xing R, Teng S, Yang Y, Zhao J, Xiao W, Zhang G, Li MJ, Yao W, Yang J. Evaluation of the efficacy and safety of immunotherapy in sarcoma: a two-center study. Front Immunol 2024; 15:1292325. [PMID: 38585276 PMCID: PMC10995229 DOI: 10.3389/fimmu.2024.1292325] [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: 09/11/2023] [Accepted: 02/21/2024] [Indexed: 04/09/2024] Open
Abstract
Background Sarcoma is a highly heterogeneous malignancy with a poor prognosis. Although chemotherapy and targeted therapy have improved the prognosis to some extent, the efficacy remains unsatisfactory in some patients. The efficacy and safety of immunotherapy in sarcoma need further evaluation. Methods We conducted a two-center study of sarcoma patients receiving PD-1 immunotherapy at Tianjin Medical University Cancer Institute and Hospital and Henan Provincial Cancer Hospital. The treatment regimens included PD-1 inhibitor monotherapy and combination therapy based on PD-1 inhibitors. The observed primary endpoints were median progression-free survival (mPFS) and median overall survival (mOS). Survival curves were compared using the Kaplan-Meier method. Results A total of 43 patients were included from the two centers. The median follow-up time for all patients was 13 months (range, 1-48 months). In the group of 37 patients with advanced or unresectable sarcoma, the mPFS was 6 months (95%CI: 5-12 months), and the mOS was 16 months (95%CI: 10-28 months). The ORR was 10.8% (4/37), and the DCR was 18.9% (7/37). Subgroup analysis showed no significant differences in mPFS (p=0.11) and mOS (p=0.88) between patients with PD-L1 negative/positive expression. There were also no significant differences in mPFS (p=0.13) or mOS (p=0.72) between PD-1 inhibitor monotherapy and combination therapy. Additionally, there were no significant differences in mPFS (p=0.52) or mOS (p=0.49) between osteogenic sarcoma and soft tissue sarcoma. Furthermore, the results showed no significant differences in mPFS (p=0.66) or mOS (p=0.96) between PD-1 inhibitors combined with targeted therapy and PD-1 inhibitors combined with AI chemotherapy. Among the 6 patients receiving adjuvant therapy after surgery, the mPFS was 15 months (95%CI: 6-NA months), and the mOS was not reached. In terms of safety, most adverse events were mild (grade 1-2) and manageable. The most severe grade 4 adverse events were bone marrow suppression, which occurred in 4 patients but resolved after treatment. There was also one case of a grade 4 adverse event related to hypertension. Conclusion Immunotherapy is an effective treatment modality for sarcoma with manageable safety. Further inclusion of more patients or prospective clinical trials is needed to validate these findings.
Collapse
Affiliation(s)
- Zhichao Liao
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jianjin Teng
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tao Li
- Department of Bone and Soft-Tissue Tumor, Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Haotian Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ting Li
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ruwei Xing
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Sheng Teng
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yun Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jun Zhao
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wanyi Xiao
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Gengpu Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weitao Yao
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
33
|
Li W, Liu L, Liang Z, Lai H, Wu J, Zhang H, Fang C. Efficacy of tyrosine kinase inhibitors in patients with advanced or metastatic sarcomas after prior chemotherapy: A meta-analysis. Medicine (Baltimore) 2024; 103:e37423. [PMID: 38489731 PMCID: PMC10939701 DOI: 10.1097/md.0000000000037423] [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: 09/21/2023] [Revised: 01/07/2024] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Sarcoma is a heterogeneous malignancy arising from interstitial tissue. Anthracycline-based therapy is the first-line treatment recommended by guidelines for patients with locally advanced or metastatic unresectable sarcoma. Recently, targeted therapies, in particular tyrosine kinase inhibitors (TKIs), have made significant progress in the treatment of sarcoma, and their efficacy has been investigated in randomized controlled trials. The aim of this meta-analysis is to evaluate the efficacy of TKIs in patients with advanced or metastatic sarcoma who have previously received chemotherapy. METHODS We completed a meta-analysis after conducting literature searches in PubMed, Embase, and Cochrane. The single-drug, placebo-controlled, randomized controlled clinical trials of TKIs in patients with advanced or progressive sarcoma who have previously received chemotherapy are available for inclusion in the study. The observation results were objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). The subgroup analysis was performed according to histological subtypes of sarcoma. RESULTS This study included 6 studies, including 1033 patients. The ORR (OR: 7.99, 95% CI: 3.62-19.61, P < .00001), DCR (OR: 2.54, 95% CI: 1.27-5.08, P = .009), PFS (HR: 0.46, 95% CI: 0.34-0.62, P < .00001), and OS (HR: 0.80, 95% CI: 0.67-0.96, P = .02) of patients treated with TKIs were better than those in the placebo group. CONCLUSIONS In patients with advanced sarcoma, TKIs have been shown to have advantages in terms of ORR, DCR and PFS and OS. Multi-targeted TKIs may be considered as one of the second-line treatment options for sarcoma patients who have received prior chemotherapy.
Collapse
Affiliation(s)
- Wenxia Li
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Liwen Liu
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Zhanpeng Liang
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Huiqin Lai
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Jiaming Wu
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Huatang Zhang
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Cantu Fang
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| |
Collapse
|
34
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
35
|
Zhang X, Xu Q, Zhang Y. Remarkable response to pazopanib plus vivolumab in a patient with pericardial synovial sarcoma carrying a novel genotype BRCA2 c.968dupT: A case report. Thorac Cancer 2024; 15:667-671. [PMID: 38323364 DOI: 10.1111/1759-7714.15237] [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: 11/30/2023] [Accepted: 01/20/2024] [Indexed: 02/08/2024] Open
Abstract
Pericardial synovial sarcomas (PSS) have a low incidence rate and are highly invasive with a dismal prognosis. Standard treatment includes surgery, radiotherapy and chemotherapy but with limited response. Here, we report the case of a 15-year-old nonsmoking youngster diagnosed with PSS who developed disease relapsed from surgery after 1 month. Next-generation sequencing (NGS) using baseline tissue was performed, and BRCA2 c.968dupT was detected. Then pazopanib (a multitargeted inhibitor) plus nivolumab (an immune checkpoint inhibitor) was administered, with a partial response and progression-free survival of 14 months. BRCA2 c.968dupT has not previously been reported in PSS and its response to targeted combination immunotherapy are not well characterized. Here, we report the efficacy of pazopanib combined with nivolumab in a PSS patient harboring BRCA2 c.968dupT and also provide the clinical evidence of the utility of NGS in exploring actionable mutations for solid tumor. Combination therapy based on immunotherapy may be a potential treatment choice for PSS harboring BRCA2 mutation.
Collapse
Affiliation(s)
- Xing Zhang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital, Department of Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, China
| | - Qinqin Xu
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Yongchang Zhang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital, Department of Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, China
| |
Collapse
|
36
|
Cho HJ, Yun KH, Shin SJ, Lee YH, Kim SH, Baek W, Han YD, Kim SK, Ryu HJ, Lee J, Cho I, Go H, Ko J, Jung I, Jeon MK, Rha SY, Kim HS. Durvalumab plus pazopanib combination in patients with advanced soft tissue sarcomas: a phase II trial. Nat Commun 2024; 15:685. [PMID: 38263321 PMCID: PMC10806253 DOI: 10.1038/s41467-024-44875-2] [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: 06/02/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
We aimed to determine the activity of the anti-VEGF receptor tyrosine-kinase inhibitor, pazopanib, combined with the anti-PD-L1 inhibitor, durvalumab, in metastatic and/or recurrent soft tissue sarcoma (STS). In this single-arm phase 2 trial (NCT03798106), treatment consisted of pazopanib 800 mg orally once a day and durvalumab 1500 mg once every 3 weeks. Primary outcome was overall response rate (ORR) and secondary outcomes included progression-free survival (PFS), overall survival, disease control rate, immune-related response criteria, and safety. The ORR was 30.4% and the trial met the pre-specified endpoint. The median PFS was 7.7 months (95% confidence interval: 5.7-10.4). The common treatment-related adverse events of grades 3-4 included neutropenia (9 [19.1%]), elevated aspartate aminotransferase (7 [14.9%]), alanine aminotransferase (5 [10.6%]), and thrombocytopenia (4 [8.5%]). In a prespecified transcriptomic analysis, the B lineage signature was a significant key determinant of overall response (P = 0.014). In situ analysis also showed that tumours with high CD20+ B cell infiltration and vessel density had a longer PFS (P = 6.5 × 10-4) than those with low B cell infiltration and vessel density, as well as better response (50% vs 12%, P = 0.019). CD20+ B cell infiltration was identified as the only independent predictor of PFS via multivariate analysis. Durvalumab combined with pazopanib demonstrated promising efficacy in an unselected STS cohort, with a manageable toxicity profile.
Collapse
Affiliation(s)
- Hee Jin Cho
- Department of Biomedical Convergence Science and Technology, CMRI, Kyungpook National University, Daegu, Republic of Korea
| | - Kum-Hee Yun
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su-Jin Shin
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Han Lee
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Wooyeol Baek
- Department of Plastic Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Dae Han
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Kyum Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyang Joo Ryu
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joohee Lee
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Iksung Cho
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Heounjeong Go
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiwon Ko
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Inkyung Jung
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Kyung Jeon
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Song Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
37
|
Dalal S, Shan KS, Thaw Dar NN, Hussein A, Ergle A. Role of Immunotherapy in Sarcomas. Int J Mol Sci 2024; 25:1266. [PMID: 38279265 PMCID: PMC10816403 DOI: 10.3390/ijms25021266] [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: 12/11/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
Sarcomas are a group of malignancies of mesenchymal origin with a plethora of subtypes. Given the sheer heterogeneity of various subtypes and the rarity of the disease, the management of sarcomas has been challenging, with poor patient outcomes. Surgery, radiation therapy and chemotherapy have remained the backbone of treatment in patients with sarcoma. The introduction of immunotherapy has revolutionized the treatment of various solid and hematological malignancies. In this review, we discuss the basics of immunotherapy and the immune microenvironment in sarcomas; various modalities of immunotherapy, like immune checkpoint blockade, oncolytic viruses, cancer-targeted antibodies, vaccine therapy; and adoptive cell therapies like CAR T-cell therapy, T-cell therapy, and TCR therapy.
Collapse
Affiliation(s)
- Shivani Dalal
- Memorial Healthcare, Division of Hematology and Oncology, Pembroke Pines, FL 33028, USA; (K.S.S.); (N.N.T.D.); (A.H.); (A.E.)
| | | | | | | | | |
Collapse
|
38
|
Walker K, Simister SK, Carr-Ascher J, Monument MJ, Thorpe SW, Randall RL. Emerging innovations and advancements in the treatment of extremity and truncal soft tissue sarcomas. J Surg Oncol 2024; 129:97-111. [PMID: 38010997 DOI: 10.1002/jso.27526] [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: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023]
Abstract
In this special edition update on soft tissue sarcomas (STS), we cover classifications, emerging technologies, prognostic tools, radiation schemas, and treatment disparities in extremity and truncal STS. We discuss the importance of enhancing local control and reducing complications, including the role of innovative imaging, surgical guidance, and hypofractionated radiation. We review advancements in systemic and immunotherapeutic treatments and introduce disparities seen in this vulnerable population that must be considered to improve overall patient care.
Collapse
Affiliation(s)
- Kyle Walker
- Department of Orthopaedics, University of California, Davis, Sacramento, California, USA
| | - Samuel K Simister
- Department of Orthopaedics, University of California, Davis, Sacramento, California, USA
| | - Janai Carr-Ascher
- Department of Hematology and Oncology, University of California, Davis, Sacramento, California, USA
| | - Michael J Monument
- Department of Surgery, The University of Calgary, Calgary, Alberta, Canada
| | - Steven W Thorpe
- Department of Orthopaedics, University of California, Davis, Sacramento, California, USA
| | - R Lor Randall
- Department of Orthopaedics, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
39
|
Jumaniyazova E, Lokhonina A, Dzhalilova D, Kosyreva A, Fatkhudinov T. Immune Cells in the Tumor Microenvironment of Soft Tissue Sarcomas. Cancers (Basel) 2023; 15:5760. [PMID: 38136307 PMCID: PMC10741982 DOI: 10.3390/cancers15245760] [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: 11/14/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Soft tissue sarcomas (STSs) are a rare heterogeneous group of malignant neoplasms characterized by their aggressive course and poor response to treatment. This determines the relevance of research aimed at studying the pathogenesis of STSs. By now, it is known that STSs is characterized by complex relationships between the tumor cells and immune cells of the microenvironment. Dynamic interactions between tumor cells and components of the microenvironment enhance adaptation to changing environmental conditions, which provides the high aggressive potential of STSs and resistance to antitumor therapy. Today, active research is being conducted to find effective antitumor drugs and to evaluate the possibility of using therapy with immune cells of STS. The difficulty in assessing the efficacy of new antitumor options is primarily due to the high heterogeneity of this group of malignant neoplasms. Studying the role of immune cells in the microenvironment in the progression STSs and resistance to antitumor therapies will provide the discovery of new biomarkers of the disease and the prediction of response to immunotherapy. In addition, it will help to initially divide patients into subgroups of good and poor response to immunotherapy, thus avoiding wasting precious time in selecting the appropriate antitumor agent.
Collapse
Affiliation(s)
- Enar Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
| | - Anastasiya Lokhonina
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Dzhuliia Dzhalilova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Anna Kosyreva
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| |
Collapse
|
40
|
Zhang R, Yang Y, Hu C, Huang M, Cen W, Ling D, Long Y, Yang XH, Xu B, Peng J, Wang S, Zhu W, Wei M, Yang J, Xu Y, Zhang X, Ma J, Wang F, Zhang H, Ma P, Zhu X, Song G, Sun LY, Wang DS, Wang FH, Li YH, Santagata S, Li Q, Feng YF, Du Z. Comprehensive analysis reveals potential therapeutic targets and an integrated risk stratification model for solitary fibrous tumors. Nat Commun 2023; 14:7479. [PMID: 37980418 PMCID: PMC10657378 DOI: 10.1038/s41467-023-43249-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
Solitary fibrous tumors (SFTs) are rare mesenchymal tumors with unpredictable evolution and with a recurrence or metastasis rate of 10-40%. Current medical treatments for relapsed SFTs remain ineffective. Here, we identify potential therapeutic targets and risk factors, including IDH1 p.R132S, high PD-L1 expression, and predominant macrophage infiltration, suggesting the potential benefits of combinational immune therapy and targeted therapy for SFTs. An integrated risk model incorporating mitotic count, density of Ki-67+ cells and CD163+ cells, MTOR mutation is developed, applying a discovery cohort of 101 primary non-CNS patients with negative tumor margins (NTM) and validated in three independent cohorts of 210 SFTs with the same criteria, and in 36 primary CNS SFTs with NTM. Compared with the existing models, our model shows significantly improved efficacy in identifying high-risk primary non-CNS and CNS SFTs with NTM for tumor progression.Our findings hold promise for advancing therapeutic strategies and refining risk prediction in SFTs.
Collapse
Affiliation(s)
- Renjing Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yang Yang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chunfang Hu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mayan Huang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wenjian Cen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Dongyi Ling
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yakang Long
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xin-Hua Yang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Boheng Xu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Junling Peng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Sujie Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Weijie Zhu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Mingbiao Wei
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jiaojiao Yang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yuxia Xu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xu Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jiangjun Ma
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Hongtu Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peiqing Ma
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaojun Zhu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Guohui Song
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Yue Sun
- Second Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - De-Shen Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Feng-Hua Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu-Hong Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Qin Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yan-Fen Feng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Ziming Du
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
41
|
Fujiwara T, Kunisada T, Nakata E, Mitsuhashi T, Ozaki T, Kawai A. Factors associated with survival in patients with clear cell sarcoma. Bone Joint J 2023; 105-B:1216-1225. [PMID: 37907082 DOI: 10.1302/0301-620x.105b11.bjj-2022-0743.r3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Aims Clear cell sarcoma (CCS) of soft-tissue is a rare melanocytic subtype of mesenchymal malignancy. The aim of this study was to investigate the clinical and therapeutic factors associated with increased survival, stratified by clinical stage, in order to determine the optimal treatment. Methods The study was a retrospective analysis involving 117 patients with histologically confirmed CCS, between July 2016 and November 2017, who were enrolled in the Bone and Soft Tissue Tumour Registry in Japan. Results The five- and ten-year survival rates were 41% (95% confidence interval (CI) 29 to 52) and 37% (95% CI 25 to 49), respectively. On multivariable analysis, the size of the tumour of > 10 cm (p = 0.006), lymph node metastasis at the time of diagnosis (p < 0.001), distant metastases at the time of diagnosis (p < 0.001), and no surgery for the primary tumour (p = 0.019) were independently associated with a poor survival. For N0M0 CCS (n = 68), the development of distant metastases was an independent prognostic factor for survival (early (< 12 months), hazard ratio (HR) 116.78 (95% CI 11.69 to 1,166.50); p < 0.001; late (> 12 months), HR 14.79 (95% CI 1.66 to 131.63); p = 0.016); neoadjuvant/adjuvant chemotherapy (p = 0.895) and/or radiotherapy (p = 0.216) were not significantly associated with survival. The five-year cumulative incidence of local recurrence was 19% (95% CI 8 to 35) and the size of the tumour was significantly associated with an increased rate of local recurrence (p = 0.012). For N1M0 CCS (n = 18), the risk of mortality was significantly lower in patients who underwent surgery for both the primary tumour and lymph node metastases (HR 0.03 (95% CI 0.00 to 0.56); p = 0.020). For M1 CCS (n = 31), excision of the primary tumour was independently associated with better survival (HR 0.26 (95% CI 0.09 to 0.76); p = 0.013). There was no significant difference in survival between the different types of systemic treatment (p = 0.523). Conclusion Complete excision of the primary tumour and lymph nodes is associated with a better survival in patients with CCS. Systemic treatment appears to provide limited benefits, demonstrating a pressing need for novel systemic agents.
Collapse
Affiliation(s)
- Tomohiro Fujiwara
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiyuki Kunisada
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Eiji Nakata
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiharu Mitsuhashi
- Centre for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Centre Hospital, Tokyo, Japan
| |
Collapse
|
42
|
Torrado C, Camaño M, Hindi N, Ortega J, Sevillano AR, Civantos G, Moura DS, Dimino A, Martín-Broto J. Antiangiogenics in Malignant Granular Cell Tumors: Review of the Literature. Cancers (Basel) 2023; 15:5187. [PMID: 37958362 PMCID: PMC10650106 DOI: 10.3390/cancers15215187] [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: 09/06/2023] [Revised: 10/12/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Granular cell tumors (GCT) represent 0.5% of all soft tissue sarcomas (STS), and when metastatic, they exhibit aggressive behavior and determine limited survival. Metastatic GCTs are relatively chemo-resistant; however, there is growing evidence of the benefit of using pazopanib and other targeted therapies in this histology. This is a review of the role of pazopanib and other targeted therapies in the treatment of GCTs, along with some insights on pathology and molecular biology described in GCTs. From 256 articles found in our search, 10 case-report articles met the inclusion criteria. Pazopanib was the most employed systemic therapy. The median reported time on therapy with pazopanib was seven months. Eight out of ten patients (80%) experienced disease control with pazopanib, while four out of ten (40%) patients achieved an objective RECIST response. Molecular studies suggested that antitumoral effects of pazopanib in GCT might be due to a loss-of-function of ATP6AP1/2 genes which consequently enhance signaling through several molecular pathways, such as SFKs, STAT5a/b, and PDGFR-β. Other reported targeted therapies for malignant GCTs included pazopanib in combination with crizotinib, which showed disease control for four months in one patient, and a PI3K inhibitor which achieved disease control for nine months in another patient. Dasatinib and megestrol were ineffective in two other different patients. Pazopanib has been demonstrated to be active in advanced GCTs and may be considered as a preferable treatment option.
Collapse
Affiliation(s)
- Carlos Torrado
- Medical Oncology Department, University Hospital Virgen del Rocío, 41013 Sevilla, Spain;
| | - Melisa Camaño
- Medical Oncology Department, National Cancer Institute, 11600 Montevideo, Uruguay;
| | - Nadia Hindi
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD), 28015 Madrid, Spain; (N.H.); (J.O.); (A.R.S.); (D.S.M.)
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040 Madrid, Spain
- General de Villalba University Hospital, 28400 Madrid, Spain
| | - Justo Ortega
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD), 28015 Madrid, Spain; (N.H.); (J.O.); (A.R.S.); (D.S.M.)
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040 Madrid, Spain
- General de Villalba University Hospital, 28400 Madrid, Spain
| | - Alberto R. Sevillano
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD), 28015 Madrid, Spain; (N.H.); (J.O.); (A.R.S.); (D.S.M.)
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040 Madrid, Spain
- General de Villalba University Hospital, 28400 Madrid, Spain
| | - Gema Civantos
- Pathology Department, Hospital Virgen del Rocío, 41013 Sevilla, Spain;
| | - David S. Moura
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD), 28015 Madrid, Spain; (N.H.); (J.O.); (A.R.S.); (D.S.M.)
| | - Alessandra Dimino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy;
| | - Javier Martín-Broto
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD), 28015 Madrid, Spain; (N.H.); (J.O.); (A.R.S.); (D.S.M.)
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040 Madrid, Spain
- General de Villalba University Hospital, 28400 Madrid, Spain
| |
Collapse
|
43
|
Rosenbaum E, Seier K, Bradic M, Kelly C, Movva S, Nacev BA, Gounder MM, Keohan ML, Avutu V, Chi P, Thornton KA, Chan JE, Dickson MA, Donoghue MT, Tap WD, Qin LX, D'Angelo SP. Immune-related Adverse Events after Immune Checkpoint Blockade-based Therapy Are Associated with Improved Survival in Advanced Sarcomas. CANCER RESEARCH COMMUNICATIONS 2023; 3:2118-2125. [PMID: 37787759 PMCID: PMC10583739 DOI: 10.1158/2767-9764.crc-22-0140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/31/2022] [Accepted: 09/26/2023] [Indexed: 10/04/2023]
Abstract
The association between immune-related AEs (irAE) and outcome in patients with sarcoma is not known. We retrospectively reviewed a cohort of patients with advanced sarcoma treated with immune checkpoint blockade (ICB)-based therapy. Association of irAEs with survival was assessed using a Cox regression model that incorporated irAE occurrence as a time-dependent covariate. Tumor samples with available RNA sequencing data were stratified by presence of an irAE to identify patterns of differential gene expression. A total of 131 patients were included. Forty-two (32%) had at least one irAE of any grade and 16 (12%) had at least one grade ≥ 3 irAE. The most common irAEs were hypothyroidism (8.3%), arthralgias (5.3%), pneumonitis (4.6%), allergic reaction (3.8%), and elevated transaminases (3.8%). Median progression-free survival (PFS) and overall survival (OS) from the time of study entry were 11.4 [95% confidence interval (CI), 10.7-15.0) and 74.6 weeks (CI, 44.9-89.7), respectively. On Cox analysis adjusting for clinical covariates that were significant in the univariate setting, the HR for an irAE (HR, 0.662; CI, 0.421-1.041) approached, but did not reach statistical significance for PFS (P = 0.074). Patients had a significantly lower HR for OS (HR, 0.443; CI, 0.246-0.798; P = 0.007) compared with those without or before an irAE. Gene expression profiling on baseline tumor samples found that patients who had an irAE had higher numbers of tumor-infiltrating dendritic cells, CD8+ T cells, and regulatory T cells as well as upregulation of immune and inflammatory pathways. SIGNIFICANCE irAE after ICB therapy was associated with an improved OS; it also approached statistical significance for improved PFS. Patients who had an irAE were more likely to have an inflamed tumor microenvironment at baseline.
Collapse
Affiliation(s)
- Evan Rosenbaum
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kenneth Seier
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martina Bradic
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ciara Kelly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sujana Movva
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Benjamin A. Nacev
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Mrinal M. Gounder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Mary L. Keohan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Viswatej Avutu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine A. Thornton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Jason E. Chan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Mark A. Dickson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Mark T.A. Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D. Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sandra P. D'Angelo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
44
|
Ikuta K, Nishida Y, Imagama S, Tanaka K, Ozaki T. The current management of clear cell sarcoma. Jpn J Clin Oncol 2023; 53:899-904. [PMID: 37451697 DOI: 10.1093/jjco/hyad083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023] Open
Abstract
Clear cell sarcoma (CCS) is a rare melanocytic soft tissue sarcoma with a high propensity for lymphatic metastasis and poor prognosis. It is characterized by the translocation of t (12;22), resulting in the rearrangement of the EWSR1 gene and overexpression of MET. Despite improvements in the diagnosis and treatment of soft tissue sarcomas, the management of CCSs remains challenging owing to their rarity, unique biological behaviour and limited understanding of their molecular pathogenesis. The standard treatment for localized CCSs is surgical excision with negative margins. However, there is an ongoing debate regarding the role of adjuvant chemotherapy, radiotherapy and lymphadenectomy in the management of this disease. CCSs are usually resistant to conventional chemotherapy. Targeted therapies, such as sunitinib and MET inhibitors, may provide promising results. Immunotherapy, particularly immune checkpoint inhibitors, is currently under investigation as a potential treatment option for CCSs. Further research is needed to better understand the biology of CCSs and develop effective therapeutic strategies. The purpose of this review is to provide a comprehensive overview of current knowledge and advances in the diagnosis and treatment of CCSs.
Collapse
Affiliation(s)
- Kunihiro Ikuta
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Yoshihiro Nishida
- Department of Rehabilitation, Nagoya University Hospital, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Shiro Imagama
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Kazuhiro Tanaka
- Department of Orthopedic Surgery, Oita University, 1-1 Idaigaoka, Hasama, Yufu City, Oita 879-5593, Japan
- Department of Advanced Medical Sciences, Oita University, 1-1 Idaigaoka, Hasama, Yufu City, Oita 879-5593, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Okayama 700-8558, Japan
| |
Collapse
|
45
|
Cohen-Nowak AJ, Dressler DB, Rock A, Mojica K, Woo D, Zuckerman LM, Chow W, Agulnik M. Role of immunotherapy in chondrosarcoma: A case report and review of the literature. Ther Adv Med Oncol 2023; 15:17588359231199877. [PMID: 37745839 PMCID: PMC10515522 DOI: 10.1177/17588359231199877] [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: 04/25/2023] [Accepted: 08/15/2023] [Indexed: 09/26/2023] Open
Abstract
Chondrosarcomas (CSs) consist of a heterogenous group of primary bone cancers arising from malignant cells which produce cartilaginous matrix. As the second most common primary bone cancer, CS are often resistant to systemic chemotherapy due to poor vascularization, slow proliferation, and expression of multidrug-resistant pumps. Immune checkpoint inhibitors have transformed the field of oncology and are now designated as frontline therapy for many solid tumor cancers. Several studies have demonstrated increased expression of programed cell death 1 (PD-1) and PD-L1 in CS tissue in vitro, which has led to the development of multiple clinical trials for immunotherapy in patients with aggressive CS. In this review, we highlight the ongoing investigation into the role for immunotherapy in CS. We also report the case of a 44-year-old female with a history of stage IV primary CS of the right shoulder who underwent radical resection with recurrence and demonstrated a spectacular sustained response to pembrolizumab at our center. Our review highlights the need for further studies investigating the role of immunotherapy in the treatment of aggressive bone sarcomas that are resistant to standard surgical resection, chemotherapy, and radiation treatment.
Collapse
Affiliation(s)
| | | | - Adam Rock
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Doni Woo
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Warren Chow
- University of California, Irvine, Irvine, CA, USA
| | - Mark Agulnik
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
46
|
Chen AP, Sharon E, O'Sullivan-Coyne G, Moore N, Foster JC, Hu JS, Van Tine BA, Conley AP, Read WL, Riedel RF, Burgess MA, Glod J, Davis EJ, Merriam P, Naqash AR, Fino KK, Miller BL, Wilsker DF, Begum A, Ferry-Galow KV, Deshpande HA, Schwartz GK, Ladle BH, Okuno SH, Beck JC, Chen JL, Takebe N, Fogli LK, Rosenberger CL, Parchment RE, Doroshow JH. Atezolizumab for Advanced Alveolar Soft Part Sarcoma. N Engl J Med 2023; 389:911-921. [PMID: 37672694 PMCID: PMC10729808 DOI: 10.1056/nejmoa2303383] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
BACKGROUND Alveolar soft part sarcoma (ASPS) is a rare soft-tissue sarcoma with a poor prognosis and no established therapy. Recently, encouraging responses to immune checkpoint inhibitors have been reported. METHODS We conducted an investigator-initiated, multicenter, single-group, phase 2 study of the anti-programmed death ligand 1 (PD-L1) agent atezolizumab in adult and pediatric patients with advanced ASPS. Atezolizumab was administered intravenously at a dose of 1200 mg (in patients ≥18 years of age) or 15 mg per kilogram of body weight with a 1200-mg cap (in patients <18 years of age) once every 21 days. Study end points included objective response, duration of response, and progression-free survival according to Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1, as well as pharmacodynamic biomarkers of multistep drug action. RESULTS A total of 52 patients were evaluated. An objective response was observed in 19 of 52 patients (37%), with 1 complete response and 18 partial responses. The median time to response was 3.6 months (range, 2.1 to 19.1), the median duration of response was 24.7 months (range, 4.1 to 55.8), and the median progression-free survival was 20.8 months. Seven patients took a treatment break after 2 years of treatment, and their responses were maintained through the data-cutoff date. No treatment-related grade 4 or 5 adverse events were recorded. Responses were noted despite variable baseline expression of programmed death 1 and PD-L1. CONCLUSIONS Atezolizumab was effective at inducing sustained responses in approximately one third of patients with advanced ASPS. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT03141684.).
Collapse
Affiliation(s)
- Alice P Chen
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Elad Sharon
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Geraldine O'Sullivan-Coyne
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Nancy Moore
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Jared C Foster
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James S Hu
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brian A Van Tine
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Anthony P Conley
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - William L Read
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Richard F Riedel
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Melissa A Burgess
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - John Glod
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Elizabeth J Davis
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Priscilla Merriam
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Abdul R Naqash
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Kristin K Fino
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brandon L Miller
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Deborah F Wilsker
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Asma Begum
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Katherine V Ferry-Galow
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Hari A Deshpande
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Gary K Schwartz
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brian H Ladle
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Scott H Okuno
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Jill C Beck
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James L Chen
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Naoko Takebe
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Laura K Fogli
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Christina L Rosenberger
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Ralph E Parchment
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James H Doroshow
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| |
Collapse
|
47
|
de Nigris F, Meo C, Palinski W. Combination of Genomic Landsscape and 3D Culture Functional Assays Bridges Sarcoma Phenotype to Target and Immunotherapy. Cells 2023; 12:2204. [PMID: 37681936 PMCID: PMC10486752 DOI: 10.3390/cells12172204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
Genomic-based precision medicine has not only improved tumour therapy but has also shown its weaknesses. Genomic profiling and mutation analysis have identified alterations that play a major role in sarcoma pathogenesis and evolution. However, they have not been sufficient in predicting tumour vulnerability and advancing treatment. The relative rarity of sarcomas and the genetic heterogeneity between subtypes also stand in the way of gaining statistically significant results from clinical trials. Personalized three-dimensional tumour models that reflect the specific histologic subtype are emerging as functional assays to test anticancer drugs, complementing genomic screening. Here, we provide an overview of current target therapy for sarcomas and discuss functional assays based on 3D models that, by recapitulating the molecular pathways and tumour microenvironment, may predict patient response to treatments. This approach opens new avenues to improve precision medicine when genomic and pathway alterations are not sufficient to guide the choice of the most promising treatment. Furthermore, we discuss the aspects of the 3D culture assays that need to be improved, such as the standardisation of growth conditions and the definition of in vitro responses that can be used as a cut-off for clinical implementation.
Collapse
Affiliation(s)
- Filomena de Nigris
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Concetta Meo
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Wulf Palinski
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA;
| |
Collapse
|
48
|
Schöffski P, Bahleda R, Wagner AJ, Burgess MA, Junker N, Chisamore M, Peterson P, Szpurka AM, Ceccarelli M, Tap WD. Results of an Open-label, Phase Ia/b Study of Pembrolizumab plus Olaratumab in Patients with Unresectable, Locally Advanced, or Metastatic Soft-Tissue Sarcoma. Clin Cancer Res 2023; 29:3320-3328. [PMID: 37382656 PMCID: PMC10472093 DOI: 10.1158/1078-0432.ccr-23-0742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 06/27/2023] [Indexed: 06/30/2023]
Abstract
PURPOSE The study evaluated safety and efficacy of olaratumab + pembrolizumab in patients with unresectable locally advanced/metastatic soft-tissue sarcoma (STS) with disease progression on standard treatment. PATIENTS AND METHODS This was open-label, multicenter, nonrandomized, phase Ia/Ib dose-escalation study followed by cohort expansion (olaratumab + pembrolizumab intravenous infusion). Primary objectives were safety and tolerability. RESULTS The majority of patients enrolled (n = 41) were female [phase Ia: 9 of 13, phase Ib/dose-expansion cohort (DEC), 17 of 28], aged < 65 years. In phases Ia and Ib, 13 and 26 patients received prior systemic therapy, respectively. Patients received olaratumab 15 mg/kg (phase Ia; cohort 1) or 20 mg/kg (phase Ia; cohort 2 and phase Ib) and pembrolizumab 200 mg (phase Ia/Ib). The median (Q1-Q3) duration of therapy (olaratumab) was 6.0 (3.0-11.9; cohort 1), 14.4 (12.4-20.9; cohort 2), and 14.0 (6.0-21.8) weeks (DEC). No dose-limiting toxicities and few grade ≥ 3 treatment-emergent adverse events [TEAE; 15 mg/kg: 2 (increased lipase); 20 mg/kg: 1 (increased lipase), 1 (colitis), 2 (diarrhea), 3 (anemia)] were reported. Two TEAEs (increased lipase) were related to study discontinuations. Twenty-one patients reported mild (grade ≤ 2) TEAEs [phase Ia, disease control rate (DCR):14.3% (1/7, cohort 1); 66.7% (4/6, cohort 2); no responses were reported; phase Ib, DCR: 53.6% (15/28); objective response rate: 21.4% (6/28; RECIST and irRECIST criteria)]. No response was observed in patients with programmed death ligand-1-positive tumors. CONCLUSIONS Antitumor activity was observed in some patients in DEC, and combination was well tolerated with manageable safety profile. Further studies are warranted to evaluate the efficacy and mechanistic impact of platelet-derived growth factor receptor inhibitors with immune checkpoint modulator coadministration.
Collapse
Affiliation(s)
- Patrick Schöffski
- University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | | | - Melissa A. Burgess
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Niels Junker
- Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | | | | | | | | | - William D. Tap
- Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
49
|
Park JA, Cheung NKV. Promise and Challenges of T Cell Immunotherapy for Osteosarcoma. Int J Mol Sci 2023; 24:12520. [PMID: 37569894 PMCID: PMC10419531 DOI: 10.3390/ijms241512520] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/30/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
The cure rate for metastatic or relapsed osteosarcoma has not substantially improved over the past decades despite the exploitation of multimodal treatment approaches, allowing long-term survival in less than 30% of cases. Patients with osteosarcoma often develop resistance to chemotherapeutic agents, where personalized targeted therapies should offer new hope. T cell immunotherapy as a complementary or alternative treatment modality is advancing rapidly in general, but its potential against osteosarcoma remains largely unexplored. Strategies incorporating immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR) modified T cells, and T cell engaging bispecific antibodies (BsAbs) are being explored to tackle relapsed or refractory osteosarcoma. However, osteosarcoma is an inherently heterogeneous tumor, both at the intra- and inter-tumor level, with no identical driver mutations. It has a pro-tumoral microenvironment, where bone cells, stromal cells, neovasculature, suppressive immune cells, and a mineralized extracellular matrix (ECM) combine to derail T cell infiltration and its anti-tumor function. To realize the potential of T cell immunotherapy in osteosarcoma, an integrated approach targeting this complex ecosystem needs smart planning and execution. Herein, we review the current status of T cell immunotherapies for osteosarcoma, summarize the challenges encountered, and explore combination strategies to overcome these hurdles, with the ultimate goal of curing osteosarcoma with less acute and long-term side effects.
Collapse
Affiliation(s)
- Jeong A Park
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Nai-Kong V. Cheung
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
50
|
Blay JY, Chevret S, Le Cesne A, Brahmi M, Penel N, Cousin S, Bertucci F, Bompas E, Ryckewaert T, Soibinet P, Boudou-Rouquette P, Saada Bouzid E, Soulie P, Valentin T, Lotz JP, Tosi D, Neviere Z, Cancel M, Ray-Coquard I, Gambotti L, Legrand F, Lamrani-Ghaouti A, Simon C, Even C, Massard C. Pembrolizumab in patients with rare and ultra-rare sarcomas (AcSé Pembrolizumab): analysis of a subgroup from a non-randomised, open-label, phase 2, basket trial. Lancet Oncol 2023; 24:892-902. [PMID: 37429302 DOI: 10.1016/s1470-2045(23)00282-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/25/2023] [Accepted: 06/08/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND Sarcoma is a heterogeneous group of diseases with few treatment options. Immunotherapy has shown little activity in studies including unselected sarcomas, but immune checkpoint blockers have shown activity in specific histotypes. We evaluated the activity of pembrolizumab in rare and ultra-rare sarcomas. METHODS AcSé Pembrolizumab is an ongoing phase 2, basket, multitumour study investigating the activity of pembrolizumab monotherapy in rare cancers. Here, we report the results obtained in patients with selected histotypes of rare sarcomas (incidence of less than one case per 1 000 000 people per year) recruited at 24 French hospitals. Key inclusion criteria were age 15 years or older, Eastern Cooperative Oncology Group performance status of 0-1, and advanced disease that was untreated and resistant to treatment. Patients were given pembrolizumab 200 mg intravenously on day 1 of every 21-day cycle for a maximum of 24 months. The primary endpoint was objective response rate at week 12 using Response Evaluation Criteria in Solid Tumours version 1.1, assessed by local investigators. The primary endpoint and safety were analysed in the intention-to-treat population. The AcSé Pembrolizumab study is registered with ClinicalTrials.gov, NCT03012620. FINDINGS Between Sept 4, 2017, and Dec 29, 2020, 98 patients were enrolled, of whom 97 received treatment and were included in analyses (median age 51 years [IQR 35-65]; 53 [55%] were male; 44 [45%] were female; no data were collected on race or ethnicity). 34 (35%) patients had chordomas, 14 (14%) had alveolar soft part sarcomas, 12 (12%) had SMARCA4-deficient sarcomas or malignant rhabdoid tumours, eight (8%) had desmoplastic small round cell tumours, six (6%) had epithelioid sarcomas, four (4%) had dendritic cell sarcomas, three (3%) each had clear cell sarcomas, solitary fibrous tumours, and myxoid liposarcomas, and ten (10%) had other ultra-rare histotypes. As of data cutoff (April 11, 2022), median follow-up was 13·1 months (range 0·1-52·8; IQR 4·3-19·7). At week 12, objective response rate was 6·2% (95% CI 2·3-13·0), with no complete responses and six partial responses in the 97 patients. The most common grade 3-4 adverse events were anaemia (eight [8%] of 97), alanine aminotransferase and aspartate aminotransferase increase (six [6%]), and dyspnoea (five [5%]). 86 serious adverse events were reported in 37 patients. Five deaths due to adverse events were reported, none of which were determined to be related to treatment (two due to disease progression, two due to cancer, and one due to unknown cause). INTERPRETATION Our data show the activity and manageable toxicity of pembrolizumab in some rare and ultra-rare sarcoma histotypes, and support the PD-1/PD-L1 pathway as a potential therapeutic target in selected histotypes. The completion of the basket study will provide further evidence regarding the activity and toxicity of pembrolizumab in identified rare types of cancer. FUNDING The Ligue contre le cancer, INCa, MSD. TRANSLATION For the French translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Jean-Yves Blay
- Centre Léon Bérard & Université Claude Bernard Lyon 1, Lyon, France.
| | - Sylvie Chevret
- Service de Biostatistique, Hôpital Saint Louis (AP-HP), Université Paris Cité, Paris, France
| | - Axel Le Cesne
- Gustave Roussy, Cancer Campus, Grand Paris, Villejuif, France
| | - Mehdi Brahmi
- Centre Léon Bérard & Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | - Emmanuelle Bompas
- Institut de Cancérologie de l'Ouest, Centre René Gauducheau, Nantes, France
| | | | | | | | | | - Patrick Soulie
- Institut de Cancérologie de l'Ouest, Centre Paul Papin, Angers, France
| | | | | | - Diego Tosi
- Institut Régional du Cancer de Montpellier, Centre Val d'Aurelle, Montpellier, France
| | | | | | | | | | | | | | | | - Caroline Even
- Gustave Roussy, Cancer Campus, Grand Paris, Villejuif, France
| | | |
Collapse
|