1
|
Graham CE, Velasco R, Alarcon Tomas A, Stewart OP, Dachy G, Del Bufalo F, Doglio M, Henter JI, Ortí G, Peric Z, Roddie C, van de Donk NWCJ, Frigault MJ, Ruggeri A, Onida F, Sánchez-Ortega I, Yakoub-Agha I, Penack O. Non-ICANS neurological complications after CAR T-cell therapies: recommendations from the EBMT Practice Harmonisation and Guidelines Committee. Lancet Oncol 2025; 26:e203-e213. [PMID: 40179916 DOI: 10.1016/s1470-2045(24)00715-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 04/05/2025]
Abstract
Neurological complications are an important concern in patients undergoing chimeric antigen receptor (CAR) T-cell therapy. Consensus guidelines inform the management of immune effector cell-associated neurotoxicity syndrome (ICANS). However, these guidelines are based on the early clinical experience with CD19 targeting CAR T cells in B-cell malignancies. In contrast, there are so far no published best practice recommendations on the current management of other non-classical neurological complications, which frequently develop after CAR T-cell infusion and cause clinically significant neurotoxicity. These non-classical neurological complications could be more prevalent because of additional CAR T-cell targets (eg, B cell maturation antigen [BCMA]), widened access, new indications in clinical development (including solid tumours in the CNS), and long-term follow-up. In this Review, the European Society for Blood and Marrow Transplantation (EBMT) Practice Harmonisation and Guidelines Committee provides recommendations on the management of CAR T-cell associated neurological complications that occur after treatment with the licensed CD19 and BCMA CAR T cells, as well as neurological toxicities that are emerging with CAR T cells in clinical trials for solid and haematological cancers. We address movement and neurocognitive toxicity, cranial nerve palsies, tumour inflammation-associated neurotoxicity, stroke, myelopathy, peripheral neuropathy, Guillain-Barré syndrome, fludarabine-associated neurotoxicity, and provide guidance on the psychological support for patients. CNS infections were excluded. The guidelines were developed based on the currently available literature and expert opinion. Recommendations are provided when possible, and areas for further research are highlighted to provide a framework to improve patient care.
Collapse
Affiliation(s)
- Charlotte E Graham
- Transplant Complications Working Party, EBMT, Paris, France; School of Cancer and Pharmaceutical Sciences, King's College London, London, UK; Department of Haematology, King's College Hospital NHS Foundation Trust, London, UK.
| | - Roser Velasco
- Department of Neurology, Neuro-oncology Unit, Institut Català d'Oncologia - Hospital Universitari de Bellvitge, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Ana Alarcon Tomas
- Cellular Therapy & Immunobiology Working Party, EBMT, Paris, France; Hematology Department, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Orla P Stewart
- Department of Haematology, King's College Hospital NHS Foundation Trust, London, UK
| | - Guillaume Dachy
- Hematology Service, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Francesca Del Bufalo
- Department of Hematology/Oncology, Cell and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico, Bambino Gesù Children's Hospital, Rome, Italy
| | - Matteo Doglio
- Autoimmune Diseases Working Party, EBMT, Paris, France; Experimental Hematology Unit, Vita-Salute University, Milan, Italy; Pediatric Immuno-Hematology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Jan-Inge Henter
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Guillermo Ortí
- Department of Hematology, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Zinaida Peric
- Transplant Complications Working Party, EBMT, Paris, France; Department of Haematology, University Hospital Centre Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Claire Roddie
- Research Department of Haematology, University College London, London, UK; Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Niels W C J van de Donk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA; Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Pathology and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Annalisa Ruggeri
- Cellular Therapy & Immunobiology Working Party, EBMT, Paris, France; Practice Harmonisation and Guidelines Committee, EBMT, Paris, France; Hematology and BMT Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Onida
- Practice Harmonisation and Guidelines Committee, EBMT, Paris, France; Haematology and BMT Unit, ASST Fatebenefratelli-Sacco, Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Isabel Sánchez-Ortega
- Practice Harmonisation and Guidelines Committee, EBMT, Paris, France; Executive Office, EBMT, Barcelona, Spain
| | - Ibrahim Yakoub-Agha
- Practice Harmonisation and Guidelines Committee, EBMT, Paris, France; Centre Hospitalier Universitaire de Lille, University of Lille, INSERM U1286, Lille, France
| | - Olaf Penack
- Department of Hematology, Oncology and Tumorimmunology, Charite University Hospital, Free University of Berlin and Humboldt University of Berlin, Berlin, Germany; National Center for Tumor Diseases, Berlin, Germany
| |
Collapse
|
2
|
Zhang J, Li Z, Guo G, Jin C, Deng M. A case of synovial sarcoma of the right mid-thigh and literature review. Oncol Rev 2024; 18:1445143. [PMID: 39758586 PMCID: PMC11695431 DOI: 10.3389/or.2024.1445143] [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: 06/06/2024] [Accepted: 11/30/2024] [Indexed: 01/07/2025] Open
Abstract
Synovial sarcoma (SS) is a rare and malignant mesenchymal neoplasm. We report a case of a 16-year-old Chinese female diagnosed with biphasic synovial sarcoma. The imaging features, surgical procedures and pathological results of the lesion were described in detail. Additionally, we conducted a review of the literature on synovial sarcoma of the thigh over the past 2 decades, identifying a total of 25 relevant case reports and summarizing the characteristics of these cases. Synovial sarcoma has a high degree of malignancy, with a high recurrence and metastasis rate, and a 5-year survival rate of 36%-76% and a 10-year survival rate of 20%-63%, so early detection of the lesion and preoperative differential diagnosis are of paramount importance in the treatment of patients.
Collapse
Affiliation(s)
| | - Zhengyi Li
- Department of Ultrasound, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | | | | | | |
Collapse
|
3
|
Kawai A, Ishihara M, Nakamura T, Kitano S, Iwata S, Takada K, Emori M, Kato K, Endo M, Matsumoto Y, Kakunaga S, Sato E, Miyahara Y, Morino K, Tanaka S, Takahashi S, Matsuo F, Matsumine A, Kageyama S, Ueda T. Safety and Efficacy of NY-ESO-1 Antigen-Specific T-Cell Receptor Gene-Transduced T Lymphocytes in Patients with Synovial Sarcoma: A Phase I/II Clinical Trial. Clin Cancer Res 2023; 29:5069-5078. [PMID: 37792433 PMCID: PMC10722137 DOI: 10.1158/1078-0432.ccr-23-1456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 10/05/2023]
Abstract
PURPOSE To determine, for patients with advanced or recurrent synovial sarcoma (SS) not suitable for surgical resection and resistant to anthracycline, the safety and efficacy of the infusion of autologous T lymphocytes expressing NY-ESO-1 antigen-specific T-cell receptor (TCR) gene and siRNA to inhibit the expression of endogenous TCR (product code: TBI-1301). PATIENTS AND METHODS Eligible Japanese patients (HLA-A*02:01 or *02:06, NY-ESO-1-positive tumor expression) received cyclophosphamide 750 mg/m2 on days -3 and -2 (induction period) followed by a single dose of 5×109 (±30%) TBI-1301 cells as a divided infusion on days 0 and 1 (treatment period). Primary endpoints were safety-related (phase I) and efficacy-related [objective response rate (ORR) by RECIST v1.1/immune-related RECIST (irRECIST); phase II]. Safety- and efficacy-related secondary endpoints were considered in both phase I/II parts. RESULTS For the full analysis set (N = 8; phase I, n = 3; phase II, n = 5), the ORR was 50.0% (95% confidence interval, 15.7-84.3) with best overall partial response in four of eight patients according to RECIST v1.1/irRECIST. All patients experienced adverse events and seven of eight patients (87.5%) had adverse drug reactions, but no deaths were attributed to adverse events. Cytokine release syndrome occurred in four of eight patients (50.0%), but all cases recovered with prespecified treatment. Immune effector cell-associated neurotoxicity syndrome, replication-competent retrovirus, and lymphocyte clonality were absent. CONCLUSIONS Adoptive immunotherapy with TBI-1301 to selectively target NY-ESO-1-positive tumor cells appears to be a promising strategy for the treatment of advanced or recurrent SS with acceptable toxicity.
Collapse
Affiliation(s)
- Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Tomoki Nakamura
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Shigehisa Kitano
- Department of Advanced Medical Development, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Shintaro Iwata
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kohichi Takada
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Emori
- Department of Orthopedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koji Kato
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Makoto Endo
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopedic Surgery, Fukushima Medical University, Fukushima, Japan
| | - Shigeki Kakunaga
- Department of Orthopaedic Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Eiichi Sato
- Department of Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yoshihiro Miyahara
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Mie, Japan
| | | | | | | | | | - Akihiko Matsumine
- Department of Orthopaedics and Rehabilitation Medicine, University of Fukui, Fukui, Japan
| | | | | |
Collapse
|
4
|
Koch C, Fleischer J, Popov T, Frontzek K, Schreiner B, Roth P, Manz MG, Unseld S, Müller AMS, Russkamp NF. Diabetes insipidus and Guillain-Barré-like syndrome following CAR-T cell therapy: a case report. J Immunother Cancer 2023; 11:e006059. [PMID: 36690387 PMCID: PMC9872508 DOI: 10.1136/jitc-2022-006059] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Immune effector cell-associated neurotoxicity syndrome (ICANS) is a common adverse event of CD19-directed chimeric antigen receptor (CAR) T cell therapy. Other neurological adverse events, however, have not methodically been described and studied. Furthermore, safety data on CAR-T cell therapy in patients with central nervous system (CNS) lymphoma remain limited. MAIN BODY We here report occurrence of a Guillain-Barré-like syndrome (GBS) and central diabetes insipidus (cDI) following tisagenlecleucel therapy for relapsed high-grade lymphoma with CNS involvement. Both complications were refractory to standard treatment of ICANS. Weakness of respiratory muscles required mechanical ventilation and tracheostomy while cDI was treated with desmopressin substitution for several weeks. Muscle-nerve biopsy and nerve conduction studies confirmed an axonal pattern of nerve damage. T cell-rich infiltrates and detection of the CAR transgene in muscle-nerve sections imply a direct or indirect role of CAR-T cell-mediated inflammation. In line with current treatment guidelines for GBS, intravenous immunoglobulin was administered and gradual but incomplete recovery was observed over the course of several months. CONCLUSIONS This case report highlights the risk of rare but severe neurological adverse events, such as acute GBS or cDI, in patients treated with CAR-T cells. It further underlines the importance of appropriate patient surveillance and systematic reporting of rare complications to eventually improve treatment.
Collapse
Affiliation(s)
- Christian Koch
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Juliane Fleischer
- Institute of Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Todor Popov
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Karl Frontzek
- Institute of Neuropathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Bettina Schreiner
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Patrick Roth
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Simone Unseld
- Institute of Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Antonia M S Müller
- Department of Transfusion Medicine and Cell Therapy, Medical University Vienna, Vienna, Austria
| | - Norman F Russkamp
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW To summarize the development of modified T-cell therapies in sarcomas and discuss relevant published and ongoing clinical trials to date. RECENT FINDINGS Numerous clinical trials are underway evaluating tumor-specific chimeric antigen receptor T cells and high affinity T-cell receptor (TCR)-transduced T cells in sarcomas. Notably, translocation-dependent synovial sarcoma and myxoid/round cell liposarcoma are the subject of several phase II trials evaluating TCRs targeting cancer testis antigens New York esophageal squamous cell carcinoma-1 (NY-ESO-1) and melanoma antigen-A4 (MAGE A4), and response rates of up to 60% have been observed for NY-ESO-1 directed, modified T cells in synovial sarcoma. Challenges posed by modified T-cell therapy include limitations conferred by HLA-restriction, non-immunogenic tumor microenvironments (TME), aggressive lymphodepletion and immune-mediated toxicities restricting coinfusion of cytokines. SUMMARY Cellular therapy to augment the adaptive immune response through delivery of modified T cells is an area of novel therapeutic development in sarcomas where a reliably expressed, ubiquitous target antigen can be identified. Therapeutic tools to improve the specificity, signaling, proliferation and persistence of modified TCRs and augment clinical responses through safe manipulation of the sarcoma TME will be necessary to harness the full potential of this approach.
Collapse
|
6
|
Shalabi H, Nellan A, Shah NN, Gust J. Immunotherapy Associated Neurotoxicity in Pediatric Oncology. Front Oncol 2022; 12:836452. [PMID: 35265526 PMCID: PMC8899040 DOI: 10.3389/fonc.2022.836452] [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: 12/15/2021] [Accepted: 01/20/2022] [Indexed: 11/30/2022] Open
Abstract
Novel immunotherapies are increasingly being employed in pediatric oncology, both in the upfront and relapsed/refractory settings. Through various mechanisms of action, engagement and activation of the immune system can cause both generalized and disease site-specific inflammation, leading to immune-related adverse events (irAEs). One of the most worrisome irAEs is that of neurotoxicity. This can present as a large spectrum of neurological toxicities, including confusion, aphasia, neuropathies, seizures, and/or death, with variable onset and severity. Earlier identification and treatment, generally with corticosteroids, remains the mainstay of neurotoxicity management to optimize patient outcomes. The pathophysiology of neurotoxicity varies across the different therapeutic strategies and remains to be elucidated in most cases. Furthermore, little is known about long-term neurologic sequelae. This review will focus on neurotoxicity seen with the most common immunotherapies used in pediatric oncology, including CAR T cell therapy, alternative forms of adoptive cell therapy, antibody therapies, immune checkpoint inhibitors, and tumor vaccines. Herein we will discuss the incidence, pathophysiology, symptomatology, diagnosis, and management strategies currently being utilized for immunotherapy-associated neurotoxicity with a focus on pediatric specific considerations.
Collapse
Affiliation(s)
- Haneen Shalabi
- National Cancer Institute, Pediatric Oncology Branch, National Institutes of Health, Bethesda, MD, United States
| | - Anandani Nellan
- National Cancer Institute, Pediatric Oncology Branch, National Institutes of Health, Bethesda, MD, United States
| | - Nirali N. Shah
- National Cancer Institute, Pediatric Oncology Branch, National Institutes of Health, Bethesda, MD, United States
| | - Juliane Gust
- Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Neurology, University of Washington, Seattle, WA, United States
| |
Collapse
|
7
|
Neurotoxicity of Tumor Immunotherapy: The Emergence of Clinical Attention. JOURNAL OF ONCOLOGY 2022; 2022:4259205. [PMID: 35087588 PMCID: PMC8789457 DOI: 10.1155/2022/4259205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 02/05/2023]
Abstract
Tumor immunotherapy brings substantial and long-term clinical benefits that can even cure tumors. However, the accumulation of evidence suggests that immunotherapy also induces severe and complex neurologic immune-related adverse events (ir-AEs) and even leads to immunotherapy-related death, which arouses the concern of clinicians. The timely and accurate identification of neurotoxicity helps clinicians detect and treat these complications early, thereby enhancing treatment efficiency and improving the prognosis of patients. At present, the mechanism of neurotoxicity caused by immunotherapy has not been completely elucidated. This paper mainly reviews the clinical features, pathogenesis, and therapeutic strategies of neurologic ir-AEs.
Collapse
|
8
|
Tang F, Tie Y, Wei YQ, Tu CQ, Wei XW. Targeted and immuno-based therapies in sarcoma: mechanisms and advances in clinical trials. Biochim Biophys Acta Rev Cancer 2021; 1876:188606. [PMID: 34371128 DOI: 10.1016/j.bbcan.2021.188606] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/04/2021] [Accepted: 08/02/2021] [Indexed: 02/08/2023]
Abstract
Sarcomas represent a distinct group of rare malignant tumors with high heterogeneity. Limited options with clinical efficacy for the metastatic or local advanced sarcoma existed despite standard therapy. Recently, targeted therapy according to the molecular and genetic phenotype of individual sarcoma is a promising option. Among these drugs, anti-angiogenesis therapy achieved favorable efficacy in sarcomas. Inhibitors targeting cyclin-dependent kinase 4/6, poly-ADP-ribose polymerase, insulin-like growth factor-1 receptor, mTOR, NTRK, metabolisms, and epigenetic drugs are under clinical evaluation for sarcomas bearing the corresponding signals. Immunotherapy represents a promising and favorable method in advanced solid tumors. However, most sarcomas are immune "cold" tumors, with only alveolar soft part sarcoma and undifferentiated pleomorphic sarcoma respond to immune checkpoint inhibitors. Cellular therapies with TCR-engineered T cells, chimeric antigen receptor T cells, tumor infiltrating lymphocytes, and nature killer cells transfer show therapeutic potential. Identifying tumor-specific antigens and exploring immune modulation factors arguing the efficacy of these immunotherapies are the current challenges. This review focuses on the mechanisms, advances, and potential strategies of targeted and immune-based therapies in sarcomas.
Collapse
Affiliation(s)
- Fan Tang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China; Department of Orthopeadics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Tie
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yu-Quan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chong-Qi Tu
- Department of Orthopeadics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Xia-Wei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Mitchell G, Pollack SM, Wagner MJ. Targeting cancer testis antigens in synovial sarcoma. J Immunother Cancer 2021; 9:jitc-2020-002072. [PMID: 34083416 PMCID: PMC8183285 DOI: 10.1136/jitc-2020-002072] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 02/02/2023] Open
Abstract
Synovial sarcoma (SS) is a rare cancer that disproportionately affects children and young adults. Cancer testis antigens (CTAs) are proteins that are expressed early in embryonic development, but generally not expressed in normal tissue. They are aberrantly expressed in many different cancer types and are an attractive therapeutic target for immunotherapies. CTAs are expressed at high levels in SS. This high level of CTA expression makes SS an ideal cancer for treatment strategies aimed at harnessing the immune system to recognize aberrant CTA expression and fight against the cancer. Pivotal clinical trials are now underway, with the potential to dramatically alter the landscape of SS management and treatment from current standards of care. In this review, we describe the rationale for targeting CTAs in SS with a focus on NY-ESO-1 and MAGE-A4, the current state of vaccine and T-cell receptor-based therapies, and consider emerging opportunities for future development.
Collapse
Affiliation(s)
| | - Seth M Pollack
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Oncology, University of Washington, Seattle, Washington, USA.,Lurie Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Michael J Wagner
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA .,Oncology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
10
|
Yang Y, Guo X, Hu B, He P, Jiang X, Wang Z, Zhu H, Hu L, Yu M, Feng M. Generated SecPen_NY-ESO-1_ubiquitin-pulsed dendritic cell cancer vaccine elicits stronger and specific T cell immune responses. Acta Pharm Sin B 2021; 11:476-487. [PMID: 33643825 PMCID: PMC7893120 DOI: 10.1016/j.apsb.2020.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/22/2022] Open
Abstract
Dendritic cell-based cancer vaccines (DC vaccines) have been proved efficient and safe in immunotherapy of various cancers, including melanoma, ovarian and prostate cancer. However, the clinical responses were not always satisfied. Here we proposed a novel strategy to prepare DC vaccines. In the present study, a fusion protein SNU containing a secretin-penetratin (SecPen) peptide, NY-ESO-1 and ubiquitin was designed and expressed. To establish the DC vaccine (DC-SNU), the mouse bone marrow-derived DCs (BMDCs) were isolated, pulsed with SNU and maturated with cytokine cocktail. Then peripheral blood mononuclear cells (PBMCs) from C57BL/6 mice inoculated intraperitoneally with DC-SNU were separated and cocultured with MC38/MC38NY-ESO-1 tumor cells or DC vaccines. The results show that SNU was successfully expressed. This strategy made NY-ESO-1 entering cytoplasm of BMDCs more efficiently and degraded mainly by proteasome. As we expected, mature BMDCs expressed higher CD40, CD80 and CD86 than immature BMDCs. Thus, the PBMCs released more IFN-γ and TNF-α when stimulated with DC-SNU in vitro again. What's more, the PBMCs induced stronger and specific cytotoxicity towards MC38NY-ESO-1 tumor cells. Given the above, it demonstrated that DC-SNU loaded with SecPen and ubiquitin-fused NY-ESO-1 could elicit stronger and specific T cell immune responses. This strategy can be used as a platform for DC vaccine preparation and applied to various cancers treatment.
Collapse
Affiliation(s)
- Yunkai Yang
- Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiaohan Guo
- Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Bo Hu
- Shanghai Novoprotein Biotechnology Co., Ltd., Shanghai 201203, China
| | - Peng He
- Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiaowu Jiang
- Medical School of Yichun University, Yichun 336000, China
| | - Zuohuan Wang
- Clinical Research Center, 2nd Affiliated Hospital, Medical College of Zhejiang University, Hangzhou 310009, China
| | - Huaxing Zhu
- Shanghai Novoprotein Biotechnology Co., Ltd., Shanghai 201203, China
| | - Lina Hu
- Department of Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medicine Center, Shanghai 201399, China
- Corresponding authors. Tel.: +86 21 51980035 (Meiqing Feng); +86 21 68035322 (Minghua Yu); +86 21 68035322 (Lina Hu).
| | - Minghua Yu
- Department of Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medicine Center, Shanghai 201399, China
- Corresponding authors. Tel.: +86 21 51980035 (Meiqing Feng); +86 21 68035322 (Minghua Yu); +86 21 68035322 (Lina Hu).
| | - Meiqing Feng
- Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Corresponding authors. Tel.: +86 21 51980035 (Meiqing Feng); +86 21 68035322 (Minghua Yu); +86 21 68035322 (Lina Hu).
| |
Collapse
|
11
|
Orcurto A, Hottinger A, Wolf B, Navarro Rodrigo B, Ochoa de Olza M, Auger A, Kuntzer T, Comte D, Zimmer V, Gannon P, Kandalaft L, Michielin O, Zimmermann S, Harari A, Trueb L, Coukos G. Guillain-Barré syndrome after adoptive cell therapy with tumor-infiltrating lymphocytes. J Immunother Cancer 2020; 8:jitc-2020-001155. [PMID: 32847987 PMCID: PMC7451492 DOI: 10.1136/jitc-2020-001155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TILs) is a promising experimental immunotherapy that has shown high objective responses in patients with melanoma. Current protocols use a lymphodepletive chemotherapy before infusion of ex vivo expanded TILs, followed by high-dose interleukin-2 (IL-2). Treatment-related toxicities are mainly attributable to the chemotherapy regimen and to the high-dose IL-2 and are generally reversible. Neurological side effects have rarely been described. Nevertheless, due to improvements in cell production techniques and due to combinations with other immunomodulating molecules, side effects not previously described may be encountered. CASE PRESENTATION We report the case of a 53-year-old heavily pretreated patient with melanoma who developed Guillain-Barré syndrome (GBS) 19 days after ACT using autologous TILs, given in the context of a phase I trial. He presented with dorsal back pain, unsteady gait and numbness in hands and feet. Lumbar puncture showed albuminocytological dissociation, and nerve conduction studies revealed prolonged distal motor latencies in median, ulnar, tibial and peroneal nerves, compatible with a GBS. The patient was treated with intravenous immunoglobulins and intensive neurological rehabilitation, with progressive and full recovery at 21 months post-TIL-ACT. Concomitant to the onset of GBS, a cytomegalovirus reactivation on immunosuppression was detected and considered as the most plausible cause of this neurological side effect. CONCLUSION We describe for the first time a case of GBS occurring shortly after TIL-ACT for melanoma, even though we could not identify with certainty the triggering agent. The report of such rare cases is of extreme importance to build on the knowledge of immune cellular therapies and their specific spectrum of toxicities.
Collapse
Affiliation(s)
- Angela Orcurto
- Immuno-oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Andreas Hottinger
- Oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland.,Service of Neurology, Department of Clinical Neurosciences, CHUV, Lausanne, Vaud, Switzerland
| | - Benita Wolf
- Oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Blanca Navarro Rodrigo
- Immuno-oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Maria Ochoa de Olza
- Immuno-oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Aymeric Auger
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Vaud, Switzerland.,Center of Experimental Therapeutics, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Thierry Kuntzer
- Service of Neurology, Department of Clinical Neurosciences, CHUV, Lausanne, Vaud, Switzerland
| | - Denis Comte
- Service of Immunology and Allergy, Department of Medicine, CHUV, Lausanne, Vaud, Switzerland
| | - Virginie Zimmer
- Center of Experimental Therapeutics, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Philippe Gannon
- Center of Experimental Therapeutics, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Lana Kandalaft
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Vaud, Switzerland.,Center of Experimental Therapeutics, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Olivier Michielin
- Oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Stefan Zimmermann
- Immuno-oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Alexandre Harari
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Vaud, Switzerland.,Center of Experimental Therapeutics, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - Lionel Trueb
- Immuno-oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland
| | - George Coukos
- Immuno-oncology Service, Department of Oncology, CHUV, Lausanne, Vaud, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Vaud, Switzerland
| |
Collapse
|