101
|
Derenzini E, Tabanelli V, Sammassimo S, Mazzara S, Motta G, Melle F, Vanazzi A, Calleri A, Fiori S, Finazzi MC, Barbanti MC, Ramadan S, Gandini S, Pastano R, Rambaldi A, Pileri S, Tarella C. Pre-existing immune checkpoints activation predicts relapse after allogeneic stem cell transplantation in lymphoma. Bone Marrow Transplant 2021; 56:2280-2283. [PMID: 33963303 DOI: 10.1038/s41409-021-01320-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Enrico Derenzini
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy. .,Department of Health Sciences, University of Milan, Milan, Italy.
| | - Valentina Tabanelli
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Simona Sammassimo
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Saveria Mazzara
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanna Motta
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Melle
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Anna Vanazzi
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Angelica Calleri
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Stefano Fiori
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Chiara Finazzi
- Hematology and Bone marrow Transplant Unit, ASST-Papa Giovanni XXIII, Bergamo, Italy
| | - Maria Chiara Barbanti
- Hematology and Bone marrow Transplant Unit, ASST-Papa Giovanni XXIII, Bergamo, Italy
| | - Safaa Ramadan
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Gandini
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Rocco Pastano
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Alessandro Rambaldi
- Hematology and Bone marrow Transplant Unit, ASST-Papa Giovanni XXIII, Bergamo, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefano Pileri
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Corrado Tarella
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy. .,Department of Health Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
102
|
Efficacy and safety of GLS-010 (zimberelimab) in patients with relapsed or refractory classical Hodgkin lymphoma: A multicenter, single-arm, phase II study. Eur J Cancer 2021; 164:117-126. [PMID: 34462189 DOI: 10.1016/j.ejca.2021.07.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/10/2021] [Accepted: 07/28/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND GLS-010 (zimberelimab) is a novel, fully human, anti-programmed death-1 monoclonal antibody that shows promising efficacy and safety in advanced solid tumors. This trial aimed to evaluate the efficacy and safety of GLS-010 (zimberelimab) in Chinese patients with relapsed or refractory classical Hodgkin lymphoma (r/r-cHL). METHODS This phase II, single-arm, open-label, multicenter clinical trial was conducted at 24 centers in China and enrolled patients with r/r-cHL after two or more lines of therapy. The patients were administered intravenous GLS-010 (zimberelimab) (240 mg, once every 2 weeks) until progression, death, unacceptable toxicity, or consent withdrawal. The primary end-point was the objective response rate assessed by an independent radiology review committee (IRC). This study was registered (NCT03655483). RESULTS Eighty-five patients were enrolled between August 2018 and August 2019. The median follow-up was 15.8 months. Seventy-seven patients (90.6%; 95% confidence interval [CI] 82.3-95.9) had an IRC-assessed objective response. The complete response rate was 32.9% (n = 28). The 12-month progression-free survival and overall survival rates were 78% (95% CI 67.5-85.6) and 99% (95% CI 91.9-99.8), respectively. Treatment-related adverse events (TRAEs) were observed in 92.9% of participants. Grade III or IV TRAEs occurred in 24 (28.2%) of the 85 participants. The most common grade III or IV TRAEs were abnormal hepatic function (5.9%), hyperuricemia (4.7%), decreased neutrophil count (3.5%), and increased weight (3.5%). Only one grade V AE, gastrointestinal infection, occurred. CONCLUSIONS GLS-010 (zimberelimab) appears to be effective and safe for the treatment of Chinese patients with r/r-cHL. Long-term follow-up is required to confirm these clinical benefits.
Collapse
|
103
|
Gao S, Yang X, Xu J, Qiu N, Zhai G. Nanotechnology for Boosting Cancer Immunotherapy and Remodeling Tumor Microenvironment: The Horizons in Cancer Treatment. ACS NANO 2021; 15:12567-12603. [PMID: 34339170 DOI: 10.1021/acsnano.1c02103] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunotherapy that harnesses the human immune system to fight cancer has received widespread attention and become a mainstream strategy for cancer treatment. Cancer immunotherapy not only eliminates primary tumors but also treats metastasis and recurrence, representing a major advantage over traditional cancer treatments. Recently with the development of nanotechnology, there exists much work applying nanomaterials to cancer immunotherapy on the basis of their excellent physiochemical properties, such as efficient tissue-specific delivery function, huge specific surface area, and controllable surface chemistry. Consequently, nanotechnology holds significant potential in improving the efficacy of cancer immunotherapy. Nanotechnology-based immunotherapy mainly manifests its inhibitory effect on tumors via two different approaches: one is to produce an effective anti-tumor immune response during tumorigenesis, and the other is to enhance tumor immune defense ability by modulating the immune suppression mechanism in the tumor microenvironment. With the success of tumor immunotherapy, understanding the interaction between the immune system and smart nanomedicine has provided vigorous vitality for the development of cancer treatment. This review highlights the application, progress, and prospect of nanomedicine in the process of tumor immunoediting and also discusses several engineering methods to improve the efficiency of tumor treatment.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Xiaoye Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Jiangkang Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Na Qiu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| |
Collapse
|
104
|
Thangam R, Patel KD, Kang H, Paulmurugan R. Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy-Current Status and Future Perspectives. Vaccines (Basel) 2021; 9:vaccines9080935. [PMID: 34452059 PMCID: PMC8402739 DOI: 10.3390/vaccines9080935] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022] Open
Abstract
Engineering polymeric nanoparticles for their shape, size, surface chemistry, and functionalization using various targeting molecules has shown improved biomedical applications for nanoparticles. Polymeric nanoparticles have created tremendous therapeutic platforms, particularly applications related to chemo- and immunotherapies in cancer. Recently advancements in immunotherapies have broadened this field in immunology and biomedical engineering, where "immunoengineering" creates solutions to target translational science. In this regard, the nanoengineering field has offered the various techniques necessary to manufacture and assemble multifunctional polymeric nanomaterial systems. These include nanoparticles functionalized using antibodies, small molecule ligands, targeted peptides, proteins, and other novel agents that trigger and encourage biological systems to accept the engineered materials as immune enhancers or as vaccines to elevate therapeutic functions. Strategies to engineer polymeric nanoparticles with therapeutic and targeting molecules can provide solutions for developing immune vaccines via maintaining the receptor storage in T- and B cells. Furthermore, cancer immunotherapy using polymeric nanomaterials can serve as a gold standard approach for treating primary and metastasized tumors. The current status of the limited availability of immuno-therapeutic drugs highlights the importance of polymeric nanomaterial platforms to improve the outcomes via delivering anticancer agents at localized sites, thereby enhancing the host immune response in cancer therapy. This review mainly focuses on the potential scientific enhancements and recent developments in cancer immunotherapies by explicitly discussing the role of polymeric nanocarriers as nano-vaccines. We also briefly discuss the role of multifunctional nanomaterials for their therapeutic impacts on translational clinical applications.
Collapse
Affiliation(s)
- Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Correspondence: (R.T.); (R.P.)
| | - Kapil D. Patel
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Correspondence: (R.T.); (R.P.)
| |
Collapse
|
105
|
Camus V, Bigenwald C, Ribrag V, Lazarovici J, Jardin F, Sarkozy C. Pembrolizumab in the treatment of refractory primary mediastinal large B-cell lymphoma: safety and efficacy. Expert Rev Anticancer Ther 2021; 21:941-956. [PMID: 34233557 DOI: 10.1080/14737140.2021.1953986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Primary mediastinal large B-cell lymphoma (PMBL) is a rare subtype of lymphoma, clinically and biologically distinct from diffuse large B-cell lymphoma (DLBCL) that shows overlapping features with classical Hodgkin lymphoma (cHL). If first-line strategies lead to 80-85% of curability, relapse occurs early with a chemo-refractory disease and a poor outcome. The presence of 9p24.1 rearrangement, conducting to the overexpression of the immune checkpoint molecules PDL1 and 2, has paved the way for immune checkpoint blockers development in these entities. Pembrolizumab, an anti PD-1 checkpoint antibody, was initially approved in solid cancer and later on in the lymphoma field in cHL.Areas covered: We summarize the biology and clinical need in PMBL, leading to the rationale for checkpoint inhibitors development, as well as pembrolizumab clinical studies in this entity. To do so, we performed a PubMed search using the terms: 'PMBCL,' 'lymphoma,' 'Immune checkpoint,' and 'Pembrolizumab.'Expert opinion: Pembrolizumab showed tolerable safety profile and efficacy data in patients with PMBL who have relapsed after, or are ineligible for autologous stem cell transplant (ASCT). Some combination strategies have shown promising preliminary results, while others are currently being conducted.
Collapse
Affiliation(s)
- Vincent Camus
- Département D'hématologie, Centre Henri Becquerel, Rouen, France
| | - Camille Bigenwald
- Département D'hématologie, Institut Gustave Roussy, Villejuif, France
| | - Vincent Ribrag
- Département D'hématologie, Institut Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique Et Des Essais Précoces (DITEP), Institut Gustave Roussy, Villejuif, France
| | - Julien Lazarovici
- Département D'hématologie, Institut Gustave Roussy, Villejuif, France
| | - Fabrice Jardin
- Département D'hématologie, Centre Henri Becquerel, Rouen, France
| | - Clémentine Sarkozy
- Département d'Innovation Thérapeutique Et Des Essais Précoces (DITEP), Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
106
|
Caro J, Diefenbach C. New approaches to managing relapsed/refractory Hodgkin lymphoma: the role of checkpoint inhibitors and beyond. Expert Rev Hematol 2021; 14:741-750. [PMID: 34350815 DOI: 10.1080/17474086.2021.1962278] [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: 10/20/2022]
Abstract
Introduction: While most patients with Hodgkin lymphoma (HL) are successfully cured with frontline therapy, unfortunately far too many patients have primary refractory disease or relapse after initial treatment, and outcomes for these patients remain suboptimal.Areas Covered: Treatment for relapsed/refractory (R/R) HL remains an ongoing challenge; however, the approval of brentuximab vedotin (BV) and the checkpoint inhibitors pembrolizumab and nivolumab have given us promising therapies with high response rates and improved progression free survival. We performed a literature search using PubMed on all HL studies investigating immunotherapy within the past 10 years.Expert Opinion: Both BV and checkpoint inhibitors have good single agent activity but appear more effective when given together and combine well with chemotherapy. Other novel agents under study include bispecific chimeric antibody constructs and chimeric antigen receptor T-cells (CAR-T). Here we review the data supporting novel therapies and immunotherapies for R/R HL.
Collapse
Affiliation(s)
- Jessica Caro
- Department of Medicine, Division of Hematology and Medical Oncology, Perlmutter Cancer Center at NYU Langone Health, New York, NY, United States
| | - Catherine Diefenbach
- Department of Medicine, Division of Hematology and Medical Oncology, Perlmutter Cancer Center at NYU Langone Health, New York, NY, United States
| |
Collapse
|
107
|
Liu Y, Guo X, Zhan L, Wang L, Wang X, Jiang M. LAG3 and PD1 Regulate CD8+ T Cell in Diffuse Large B-cell Lymphoma Patients. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:4468140. [PMID: 34422089 PMCID: PMC8378962 DOI: 10.1155/2021/4468140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/17/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is a clinically and genetically heterogeneous lymphoid malignancy. The unsatisfactory outcome for refractory patients has prompted efforts to explore new therapeutic approaches for DLBCL. However, the mechanisms involved in treatment associated with immune checkpoints remain unclear. This study is aimed at investigating the potential roles of programmed cell death protein 1 (PD1) and lymphocyte activation gene 3 (LAG3) in CD8+ T cells for treatment in DLBCL. METHODS Utilizing flow cytometry, we examined the content of T cells, the levels of cytokines, and the expression of PD1 and LAG3 in patients with DLBCL as well as in healthy controls. Levels of cytokines in CD8+ T cells from DLBCL patients before and after treatment were compared by blocking of PD1 and LAG3 in magnetic bead-sorted CD8+ T cells. RESULTS We found that the proportion of CD4+ T cells and CD8+ T cells was increased in DLBCL patients after treatment. The levels of cytokines trended toward those of healthy controls in treatment. PD1 (+), LAG3 (+), or PD1 (+) LAG3 (+) were all expressed in lower amounts in CD4+ T cells and CD8+ T cells after treatment than in untreated DLBCL patients. In addition, blockade of PD1 and LAG3 in sorted CD8+ T cells markedly inhibited cytokine production in response to treatment. CONCLUSION PD1 and LAG3 in CD8+ T cells may be important targets of therapy and play therapeutic role in patients with DLBCL.
Collapse
Affiliation(s)
- Ying Liu
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang, China
- Xinjiang Uygur Autonomous Region Research Institute of Hematology, Urumqi, 830061 Xinjiang, China
| | - Xinhong Guo
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang, China
- Xinjiang Uygur Autonomous Region Research Institute of Hematology, Urumqi, 830061 Xinjiang, China
| | - Lingbo Zhan
- Xinjiang Medical University, Urumqi, 830000 Xinjiang, China
| | - Lei Wang
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang, China
- Xinjiang Uygur Autonomous Region Research Institute of Hematology, Urumqi, 830061 Xinjiang, China
| | - Xinyou Wang
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang, China
- Xinjiang Uygur Autonomous Region Research Institute of Hematology, Urumqi, 830061 Xinjiang, China
| | - Ming Jiang
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang, China
- Xinjiang Uygur Autonomous Region Research Institute of Hematology, Urumqi, 830061 Xinjiang, China
| |
Collapse
|
108
|
Nigro O, Ferrari A, Casanova M, Orbach D, Leruste A, Gatz SA, Frappaz D, Massimino M. Controversies on the possible role of immune checkpoint inhibitors in pediatric cancers: balancing irAEs and efficacy. TUMORI JOURNAL 2021; 107:276-281. [PMID: 33877022 DOI: 10.1177/03008916211010214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pediatric cancers are not the equivalent of adult cancers occurring at a younger age and the prospect of immunotherapy in children has not been received with the same enthusiasm as in the adult setting. Although most pediatric malignancies are considered immunologically cold, we are learning more about PD-L1 expression, tumor mutational burden, and microsatellite instability in several pediatric cancers. The side effects of immunotherapy are an important consideration. Immune checkpoint inhibitors (ICIs) engender a unique constellation of inflammatory toxicities known as immune-related adverse events (irAEs). Three early-phase trials-KEYNOTE-051, iMATRIX, and ADVL1412-were the first to describe irAEs in pediatric patients and ICIs were well tolerated. There was concern about unknown late irAEs in pediatric patients, as they have much more time to develop than in adult or elderly patients. Academic clinicians, biopharmaceutical companies, and parents' advocates concluded that no benefit could be expected from further monotherapy trials employing other ICIs with the same mechanism of action until more scientific knowledge becomes available. On the other hand, ICIs could be useful in combination with other therapies to prevent the functional inactivation of several pathways in the hostile microenvironment. Future clinical studies on ICIs in children need to build on strong biological premises, taking into account the distinctive immunobiology of pediatric cancers vis-à-vis ICI-responsive adult cancers. We need to gain and share experiences of new therapies for managing pediatric patients with cancer, clarifying to what extent we can extrapolate the data obtained in adults.
Collapse
Affiliation(s)
- Olga Nigro
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Andrea Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Michela Casanova
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Daniel Orbach
- SIREDO Pediatric Cancer Center, Institut Curie, PSL Research University, Paris, France
| | - Amaury Leruste
- SIREDO Pediatric Cancer Center, Institut Curie, PSL Research University, Paris, France
| | - Susanne A Gatz
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Didier Frappaz
- Département de Neuro-Oncologie, Centre Léon-Bérard, Institut d'Hématologie et Oncologie Pédiatrique et Adulte, Lyon, France
| | - Maura Massimino
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| |
Collapse
|
109
|
Immune checkpoint inhibitors and allogeneic transplant in lymphoid malignancies: a deceptive friend story. Bone Marrow Transplant 2021; 56:2624-2625. [PMID: 34304253 DOI: 10.1038/s41409-021-01396-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 11/08/2022]
|
110
|
Pilkington P, Lopci E, Adam JA, Kobe C, Goffin K, Herrmann K. FDG-PET/CT Variants and Pitfalls in Haematological Malignancies. Semin Nucl Med 2021; 51:554-571. [PMID: 34272037 DOI: 10.1053/j.semnuclmed.2021.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hematologic malignancies represent a vast group of hematopoietic and lymphoid cancers that typically involve the blood, the bone marrow, and the lymphatic organs. Due to extensive research and well defined and standardized response criteria, the role of [18F]FDG-PET/CT is well defined in these malignancies. Never the less, the reliability of visual and quantitative interpretation of PET/CT may be impaired by several factors including inconsistent scanning protocols and image reconstruction methods. Furthermore, the uptake of [18F]FDG not only reflects tissue glucose consumption by malignant lesions, but also in other situations such as in inflammatory lesions, local and systemic infections, benign tumors, reactive thymic hyperplasia, histiocytic infiltration, among others; or following granulocyte colony stimulating factors therapy, radiation therapy, chemotherapy or surgical interventions, all of which are a potential source of false-positive or negative interpretations. Therefore it is of paramount importance for the Nuclear Medicine Physician to be familiar with, not only the normal distribution of [18F]FDG in the body, but also with the most frequent findings that may hamper a correct interpretation of the scan, which could ultimately alter the patients management. In this review, we describe these myriad of situations so the interpreting physician can be familiar with them, providing tools for their correct identification and interpretation when possible.
Collapse
Affiliation(s)
- Patrick Pilkington
- Department of Nuclear Medicine, University Hospital 12 de Octubre, Madrid, Spain.
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS-Humanitas Research Hospital, Rozzano (Milano), Italy
| | - Judit A Adam
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Karolien Goffin
- Department of Nuclear Medicine, University Hospital Leuven, Division of Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen Germany; West German Cancer Center
| |
Collapse
|
111
|
Tarekegn K, Colon Ramos A, Singh B, Sequeira Gross HG, Gupta S. Checkpoint Inhibitors in Relapsed/Refractory Classical Hodgkin Lymphoma. World J Oncol 2021; 12:81-84. [PMID: 34349851 PMCID: PMC8297052 DOI: 10.14740/wjon1388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Even though classical Hodgkin lymphoma is highly curable, the outcome of patients with a refractory or relapsed disease has been disappointing. Multiple lines of therapy are available for patients after their first failure, and most respond to subsequent therapies. However, there is a sizable proportion that remains relapsing/recurrent even after several lines of therapy. The overall prognosis of patients with relapsing and recurrent classical Hodgkin lymphoma (rrcHL) has been very disappointing until recently. Immune checkpoint inhibitors such as the anti-programmed death 1 (PD-1) receptor antibodies have recently been approved to treat relapsed and refractory cHL and have significantly improved the outcome of patients with rrcHL. The approved immune checkpoint inhibitors for relapsed and refractory cHL are nivolumab and pembrolizumab. In the Checkmate 205 study nivolumab demonstrated an objective response rate of 69% with an acceptable safety profile. Similarly, pembrolizumab demonstrated an overall response rate (ORR) of 69% with a complete remission rate (CRR) of 22.4% in the KEYNOTE-087 study in heavily pretreated patients with rrcHL.
Collapse
Affiliation(s)
- Kidist Tarekegn
- Department of Internal Medicine, St. Barnabas Hospital, Bronx, NY 10457, USA.,These authors contributed equally to this work
| | - Ana Colon Ramos
- Department of Internal Medicine, St. Barnabas Hospital, Bronx, NY 10457, USA.,These authors contributed equally to this work
| | - Balraj Singh
- Department of Hematology/Oncology, Saint Joseph's University Medical Center, Paterson, NJ 07503, USA
| | | | - Sachin Gupta
- Hospital Medicine, Tower Health Reading Hospital, West Reading, PA 19611, USA
| |
Collapse
|
112
|
Wienand K, Chapuy B. Molecular classification of aggressive lymphomas-past, present, future. Hematol Oncol 2021; 39 Suppl 1:24-30. [PMID: 34105819 DOI: 10.1002/hon.2847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Indexed: 12/12/2022]
Abstract
Aggressive large B-cell lymphomas (LBCLs) represent a frequent but clinically and molecularly heterogeneous group of tumors. Technological advances over the last decades prompted the development of different classification schemas to either sharpen diagnoses, dissect molecular heterogeneity, predict outcome, or identify rational treatment targets. Despite increased diagnostic precision and a noticeably improved molecular understanding of these lymphomas, clinical perspectives of patients largely remain unchanged. Recently, finished comprehensive genomic studies discovered genetically defined LBCL subtypes that predict outcome, provide insight into lymphomagenesis, and suggest rational therapies with the hope of generating patient-tailored treatments with increased perspective for patients in greatest need. Current and future efforts integrate multiomics studies and/or leverage single-cell technologies and will provide us with an even more fine-grained picture of LBCL biology. Here, we highlight examples of how high-throughput technologies aided in a better molecular understanding of LBCLs and provide examples of how to select rationally designed targeted treatment approaches that might personalize LBCL treatment and eventually improve patients' perspective in the near future.
Collapse
Affiliation(s)
- Kirsty Wienand
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Björn Chapuy
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
113
|
Bergamini C, Ferris RL, Xie J, Mariani G, Ali M, Holmes WC, Harrington K, Psyrri A, Cavalieri S, Licitra L. Bleeding complications in patients with squamous cell carcinoma of the head and neck. Head Neck 2021; 43:2844-2858. [PMID: 34117666 PMCID: PMC8453784 DOI: 10.1002/hed.26772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/17/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
Hemorrhage in recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) may be attributed to chemotherapy and local tumor irradiation. Evidence of the relationship between hemorrhage in R/M HNSCC and targeted therapies, including epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) inhibitors, or immune checkpoint inhibitors, is limited. We aimed to identify epidemiological and clinical data related to the occurrence of hemorrhage in R/M HNSCC and to explore its relationship with various therapies. We describe information obtained from literature searches as well as data extracted from a commercial database and a database from the author's institution (Istituto Nazionale dei Tumori of Milan). Evidence suggests that most bleeding events in R/M HNSCC are minor. Clinical trial safety data do not identify a causal association between hemorrhage and anti‐EGFR agents or immune checkpoint inhibitors. In contrast, anti‐VEGF agents are associated with increased, and often severe/fatal, hemorrhagic complications.
Collapse
Affiliation(s)
- Cristiana Bergamini
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Robert L Ferris
- Department of Otolaryngology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jing Xie
- Department of Epidemiology, AstraZeneca, Gaithersburg, Maryland, USA
| | | | - Muzammil Ali
- Global Medicine Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - William C Holmes
- Global Medicine Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Kevin Harrington
- The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, National Institute of Health Research Biomedical Research Centre, London, UK
| | - Amanda Psyrri
- Section of Medical Oncology, Department of Internal Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Italy
| |
Collapse
|
114
|
Koks MS, Ocak G, Suelmann BBM, Hulsbergen-Veelken CAR, Haitjema S, Vianen ME, Verhaar MC, Kaasjager KAH, Khairoun M. Immune checkpoint inhibitor-associated acute kidney injury and mortality: An observational study. PLoS One 2021; 16:e0252978. [PMID: 34101756 PMCID: PMC8186792 DOI: 10.1371/journal.pone.0252978] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/26/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors, approved for the treatment of various types of cancer, are known to cause a unique spectrum of side effects, including acute kidney injury (AKI). The aim of this study was to describe the incidence, risk factors, renal outcomes, and mortality of AKI in patients receiving checkpoint inhibitors. METHODS Patients receiving checkpoint inhibitors between January 2013 and May 2020 at the University Medical Center Utrecht, the Netherlands, were identified using the Utrecht Patient Oriented Database. AKI was defined as an increase in serum creatinine of ≥1.5 times the baseline value, based on the Kidney Disease: Improving Global Outcomes criteria. Cox proportional hazard regression analysis was used to assess risk factors for AKI and to evaluate the relationship between AKI and mortality. Persistent renal dysfunction was diagnosed in AKI patients with a final serum creatinine measurement of >1.3 times the baseline value. RESULTS Among 676 patients receiving checkpoint inhibitors, the overall incidence of AKI was 14.2%. Baseline variables independently associated with AKI were a gynecologic malignancy, monotherapy with ipilimumab, and the use of a diuretic, angiotensin-converting enzyme inhibitor or angiotensin-receptor blocker, or proton pump inhibitor at baseline. AKI was checkpoint inhibitor-associated in one third of all patients with AKI. Checkpoint inhibitor-associated AKI was mostly low-grade, occurred a median of 15 weeks after checkpoint inhibitor initiation, and resulted in persistent renal dysfunction in approximately 40% of the patients. Patients with all-cause AKI had a twofold increased mortality risk, but checkpoint inhibitor-associated AKI was not associated with increased mortality. CONCLUSIONS In this study, patients receiving checkpoint inhibitors frequently developed AKI due to various etiologies. AKI directly related to the effect of checkpoint inhibitor toxicity did not increase mortality. However, AKI not related to the effect of checkpoint inhibitor toxicity was associated with increased mortality.
Collapse
Affiliation(s)
- Marije S. Koks
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gurbey Ocak
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Britt B. M. Suelmann
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Saskia Haitjema
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marieke E. Vianen
- Department of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Karin A. H. Kaasjager
- Department of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Meriem Khairoun
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
115
|
Pezeshki PS, Mahdavi Sharif P, Rezaei N. Resistance mechanisms to programmed cell death protein 1 and programmed death ligand 1 inhibitors. Expert Opin Biol Ther 2021; 21:1575-1590. [PMID: 33984254 DOI: 10.1080/14712598.2021.1929919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: In the past few years, administrating monoclonal humanized antibodies, namely checkpoint inhibitors, against programmed cell death protein 1 (PD-1), and its ligand (PD-L1), has yielded reassuring tumor regression rates. Anti-PD-1/PD-L1 checkpoint inhibitors disrupt the engagement of PD-1 on T-cells and their ligands on tumor or other target cells and reactivate the tumor-specific T infiltrating lymphocytes (TILs), which are mostly in a state of anergy before the PD-1/PD-L1 blockade. However, a limited number of patients initially respond, and the others show a primary (innate) resistance. Moreover, the rate of relapse and tumor progression after a partial, or even complete response (secondary or acquired resistance) is relatively considerable.Areas covered: This paper presents a comprehensive discussion on the mechanisms of primary and secondary resistance to PD-1/PD-L1 blockade. Loss of T-cell infiltration or T-cell exclusion, lack of PD-L1 or PD-1 expression, and also lack of tumor immunogenicity are among the most important mechanisms, and also biomarkers of resistance in patients undergoing PD-1/PD-L1 blockade. Several somatic mutations in tumors are known to be related to at least one of the resistance mechanisms.Expert opinion: Identification of the novel resistance mechanisms suggests further combinatorial therapies to tackle primary and secondary resistance to PD-1/PD-L1 blockade.
Collapse
Affiliation(s)
- Parmida Sadat Pezeshki
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pouya Mahdavi Sharif
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Sheffield, UK
| |
Collapse
|
116
|
Hamadani M, Collins GP, Caimi PF, Samaniego F, Spira A, Davies A, Radford J, Menne T, Karnad A, Zain JM, Fields P, Havenith K, Cruz HG, He S, Boni J, Feingold J, Wuerthner J, Horwitz S. Camidanlumab tesirine in patients with relapsed or refractory lymphoma: a phase 1, open-label, multicentre, dose-escalation, dose-expansion study. LANCET HAEMATOLOGY 2021; 8:e433-e445. [PMID: 34048682 DOI: 10.1016/s2352-3026(21)00103-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Novel approaches are required to improve outcomes in relapsed or refractory classical Hodgkin lymphoma and non-Hodgkin lymphoma. We aimed to evaluate camidanlumab tesirine, an anti-CD25 antibody-drug conjugate, in this patient population. METHODS This was a phase 1, dose-escalation (part 1), dose-expansion (part 2), multicentre trial done in 12 hospital sites (seven in the USA and five in the UK). Adults (≥18 years old) with pathologically confirmed relapsed or refractory classical Hodgkin lymphoma or non-Hodgkin lymphoma, an Eastern Cooperative Oncology Group performance status 0-2, who had no therapies available to them with established clinical benefit for their disease stage were enrolled. Camidanlumab tesirine was administered intravenously (3-150 μg/kg) once every 3 weeks. Primary objectives were to assess dose-limiting toxicity, determine maximum tolerated dose and recommended expansion dose(s), and assess safety of camidanlumab tesirine. Safety was assessed in all treated patients; antitumour activity was assessed in patients with one or more valid baseline and post-baseline disease assessment and in those who had disease progression or died after first study-drug dose. This trial was registered with ClinicalTrials.gov, NCT02432235. FINDINGS Between Oct 5, 2015, and Jun 30, 2019, 133 patients were enrolled (77 [58%] had classical Hodgkin lymphoma and 56 (42%) had non-Hodgkin lymphoma). Median follow-up was 9·2 months (IQR 4·2-14·3). Eight dose-limiting toxicities were reported in five (6%) of 86 patients who were evaluable; the maximum tolerated dose was not reached. The recommended doses for expansion were 30 μg/kg and 45 μg/kg for patients with classical Hodgkin lymphoma and 80 μg/kg for patients with T-cell non-Hodgkin lymphomas. No recommended doses for expansion were defined for B-cell non-Hodgkin lymphomas. Grade 3 or worse treatment-emergent adverse events (reported by ≥10% of the 133 patients) included increased γ-glutamyltransferase (20 [15%] patients), maculopapular rash (16 [12%]), and anaemia (15 [11%]); 74 (56%) patients had serious treatment-emergent adverse events, most commonly pyrexia (16 [12%]). One (1%) fatal treatment-emergent adverse event and two (2%) deaths outside the reporting period were considered at least possibly study-drug related. Antitumoural activity was seen in classical Hodgkin and non-Hodgkin lymphomas; notably in all patients with classical Hodgkin lymphoma, the overall response was 71% (95% CI 60-81). INTERPRETATION These results warrant evaluation of camidanlumab tesirine as a potential treatment option for relapsed or refractory lymphoma, particularly in patients with classical Hodgkin lymphoma. FUNDING ADC Therapeutics.
Collapse
Affiliation(s)
- Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Graham P Collins
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, UK
| | - Paolo F Caimi
- Case Western Reserve University-University Hospitals Cleveland Medical Center, OH, USA
| | - Felipe Samaniego
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander Spira
- Virginia Cancer Specialists Research Institute, Fairfax, VA, USA; Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrew Davies
- Cancer Research UK and National Institute for Health Research Experimental Cancer Medicines Centre, University of Southampton, Southampton, UK
| | - John Radford
- National Institute for Health Research Manchester Clinical Research Facility, Manchester Academic Health Science Centre, University of Manchester and the Christie NHS Foundation Trust, Manchester, UK
| | - Tobias Menne
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Anand Karnad
- Cancer Therapy and Research Center, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Jasmine M Zain
- Comprehensive Cancer Center, City of Hope Duarte, Duarte, CA, USA
| | - Paul Fields
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | | | - Shui He
- ADC Therapeutics, Murray Hill, NJ, USA
| | | | | | | | - Steven Horwitz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
117
|
Hatic H, Sampat D, Goyal G. Immune checkpoint inhibitors in lymphoma: challenges and opportunities. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1037. [PMID: 34277837 PMCID: PMC8267255 DOI: 10.21037/atm-20-6833] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are immunomodulatory antibodies that intensify the host immune response, thereby leading to cytotoxicity. The primary targets for checkpoint inhibition have included cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death receptor-1 (PD-1) or programmed cell death ligand-1 (PD-L1). ICIs have resulted in a change in treatment landscape of various neoplasms. Among hematologic malignancies, ICIs have been most successful in certain subtypes of lymphomas such as classic Hodgkin lymphoma (cHL) and primary mediastinal B-cell lymphoma (PMBCL). However, there have been several challenges in harnessing the host immune system through ICI use in other lymphomas. The underlying reasons for the low efficacy of ICI monotherapy in most lymphomas may include defects in antigen presentation, non-inflamed tumor microenvironment (TME), immunosuppressive metabolites, genetic factors, and an overall lack of predictive biomarkers of response. In this review, we outline the existing and ongoing studies utilizing ICI therapy in various lymphomas. We also describe the challenges leading to the lack of efficacy with ICI use and discuss potential strategies to overcome those challenges including: chimeric antigen receptor T-cell therapy (CAR-T therapy), bispecific T-cell therapy (BiTE), lymphocyte activation gene-3 (LAG-3) inhibitors, T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) inhibitors, vaccines, promotion of inflammatory macrophages, indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors, DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi). Tumor mutational burden and interferon-gamma release assays are potential biomarkers of ICI treatment response beyond PD-L1 expression. Further collaborations between clinicians and scientists are vital to understand the immunopathology in ICI therapy in order to improve clinical outcomes.
Collapse
|
118
|
Autologous stem cell transplantation after anti-PD-1 therapy for multiply relapsed or refractory Hodgkin lymphoma. Blood Adv 2021; 5:1648-1659. [PMID: 33710337 DOI: 10.1182/bloodadvances.2020003556] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/10/2021] [Indexed: 01/14/2023] Open
Abstract
Autologous stem cell transplantation (ASCT) can be curative for patients with relapsed/refractory Hodgkin lymphoma (HL). Based on studies suggesting that anti-PD-1 monoclonal antibodies (mAbs) can sensitize patients to subsequent chemotherapy, we hypothesized that anti-PD-1 therapy before ASCT would result in acceptable outcomes among high-risk patients who progressed on or responded insufficiently to ≥1 salvage regimen, including chemorefractory patients who are traditionally considered poor ASCT candidates. We retrospectively identified 78 HL patients who underwent ASCT after receiving an anti-PD-1 mAb (alone or in combination) as third-line or later therapy across 22 centers. Chemorefractory disease was common, including 42 patients (54%) refractory to ≥2 consecutive systemic therapies immediately before anti-PD-1 treatment. Fifty-eight (74%) patients underwent ASCT after anti-PD-1 treatment, while 20 patients (26%) received additional therapy after PD-1 blockade and before ASCT. Patients received a median of 4 systemic therapies (range, 3-7) before ASCT, and 31 patients (41%) had a positive pre-ASCT positron emission tomography (PET) result. After a median post-ASCT follow-up of 19.6 months, the 18-month progression-free survival (PFS) and overall survival were 81% (95% CI, 69-89) and 96% (95% confidence interval [CI], 87-99), respectively. Favorable outcomes were observed for patients who were refractory to 2 consecutive therapies immediately before PD-1 blockade (18-month PFS, 78%), had a positive pre-ASCT PET (18-month PFS, 75%), or received ≥4 systemic therapies before ASCT (18-month PFS, 73%), while PD-1 nonresponders had inferior outcomes (18-month PFS, 51%). In this high-risk cohort, ASCT after anti-PD-1 therapy was associated with excellent outcomes, even among heavily pretreated, previously chemorefractory patients.
Collapse
|
119
|
Kunou S, Shimada K, Takai M, Sakamoto A, Aoki T, Hikita T, Kagaya Y, Iwamoto E, Sanada M, Shimada S, Hayakawa F, Oneyama C, Kiyoi H. Exosomes secreted from cancer-associated fibroblasts elicit anti-pyrimidine drug resistance through modulation of its transporter in malignant lymphoma. Oncogene 2021; 40:3989-4003. [PMID: 33994542 PMCID: PMC8195743 DOI: 10.1038/s41388-021-01829-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment is deeply involved in the process of tumor growth and development. In this study, we focused on cancer-associated fibroblasts (CAFs) and their derived exosomes on the lymphoma microenvironment to uncover their clinical significance. CAFs were established from primary lymphoma samples, and exosomes secreted from CAFs were obtained by standard procedures. We then investigated the roles of CAFs and their derived exosomes in the survival and drug resistance of lymphoma cells. CAFs supported the survival of lymphoma cells through increased glycolysis, and the extent differed among CAFs. Exosomes were identified as a major component of the extracellular vesicles from CAFs, and they also supported the survival of lymphoma cells. The suppression of RAB27B, which is involved in the secretion of exosomes, using a specific siRNA resulted in reduced exosome secretion and decreased survival of lymphoma cells. Moreover, anti-pyrimidine drug resistance was induced in the presence of exosomes through the suppression of the pyrimidine transporter, equilibrative nucleoside transporter 2 (ENT2), and the suppression of ENT2 was significant in in vivo experiments and clinical samples. RNA sequencing analysis of miRNAs in exosomes identified miR-4717-5p as one of the most abundant miRNAs in the exosome, which suppressed the expression of ENT2 and induced anti-pyrimidine drug resistance in vitro. Our results suggest that exosomes including miR-4717-5p secreted from CAFs play a pivotal role in the lymphoma microenvironment, indicating that they are a promising therapeutic target.
Collapse
Affiliation(s)
- Shunsuke Kunou
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuyuki Shimada
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Mika Takai
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.,Fujii Memorial Research Institute, Otsuka Pharmaceutical Co., Ltd, Otsu, Shiga, Japan
| | - Akihiko Sakamoto
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tomohiro Aoki
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tomoya Hikita
- Division of Cancer Cell Regulation, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan.,Department of Target and Drug Discovery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yusuke Kagaya
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Eisuke Iwamoto
- Department of Advanced Diagnosis, Clinical Research Centre, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Masashi Sanada
- Department of Advanced Diagnosis, Clinical Research Centre, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Satoko Shimada
- Department of Pathology and Clinical Laboratories, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Fumihiko Hayakawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Chitose Oneyama
- Division of Cancer Cell Regulation, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan.,Department of Target and Drug Discovery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
120
|
Fakhri B, Yilmaz E, Gao F, Ambinder RF, Jones R, Bartlett NL, Cashen A, Wagner-Johnston N. Survival after autologous versus allogeneic transplantation in patients with relapsed and refractory Hodgkin lymphoma. Leuk Lymphoma 2021; 62:2408-2415. [PMID: 33988071 DOI: 10.1080/10428194.2021.1927016] [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: 10/21/2022]
Abstract
For relapsed Hodgkin lymphoma, salvage chemotherapy followed by auto-HCT is the standard of care. It is important to identify subpopulations who could benefit from allo-HCT. This retrospective analysis included 277 patients with rrHL who underwent first transplant with auto-HCT or allo-HCT between 2007-2017. Patients in the auto-HCT cohort (N = 218) were older, more likely to be in CR at the time of transplant and receive maintenance therapy post-transplant. Patients who underwent allo-HCT (N = 59) had a higher MSKCC relapse score. Factors associated with an inferior PFS and OS included early relapse, advanced stage, extranodal involvement and not achieving CR following salvage chemotherapy. After controlling for these 4 risk factors and MSKCC score, PFS (p = 0.112) or OS (p = 0.256) was not affected by the choice of transplant. In patients with ≥ 3 high risk features, the 4-year PFS was 51% in the allo-HCT vs. 39% (p = 0.107) in the auto-HCT cohort.
Collapse
Affiliation(s)
- Bita Fakhri
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Elif Yilmaz
- Division of Hematology and Oncology, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard F Ambinder
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard Jones
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nancy L Bartlett
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Amanda Cashen
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nina Wagner-Johnston
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
121
|
KEYNOTE-013 4-year follow-up of pembrolizumab in classical Hodgkin lymphoma after brentuximab vedotin failure. Blood Adv 2021; 4:2617-2622. [PMID: 32556281 DOI: 10.1182/bloodadvances.2019001367] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/15/2020] [Indexed: 12/25/2022] Open
Abstract
The KEYNOTE-013 study was conducted to evaluate pembrolizumab monotherapy in hematologic malignancies; classical Hodgkin lymphoma (cHL) was an independent expansion cohort. We present long-term results based on >4 years of median follow-up for the cHL cohort. The trial enrolled cHL patients who experienced relapse after, were ineligible for, or declined autologous stem cell transplantation and experienced progression with or did not respond to brentuximab vedotin. Patients received IV pembrolizumab 10 mg/kg every 2 weeks for up to 2 years or until confirmed progression or unacceptable toxicity. Primary end points were safety and complete response (CR) rate by central review. Enrolled patients (N = 31) had received a median of 5 therapies (range, 2 to 15). After a median follow-up of 52.8 months (range, 7.0 to 57.6 months), CR rate was 19%, and median duration of response (DOR) was not reached; 24-month and 36-month DOR rates were both 50% by the Kaplan-Meier method. Median overall survival was not reached; 36-month overall survival was 81%. Six patients (19%) experienced grade 3 treatment-related adverse events (AEs); there were no grade 4 or 5 treatment-related AEs. With long-term follow-up among a heavily pretreated cohort, pembrolizumab had a favorable safety profile; some patients maintained long-term response with pembrolizumab years after end of treatment. This trial was registered at www.clinicaltrials.gov as #NCT01953692.
Collapse
|
122
|
Ouyang T, Cao Y, Kan X, Chen L, Ren Y, Sun T, Yan L, Xiong B, Liang B, Zheng C. Treatment-Related Serious Adverse Events of Immune Checkpoint Inhibitors in Clinical Trials: A Systematic Review. Front Oncol 2021; 11:621639. [PMID: 34046338 PMCID: PMC8144509 DOI: 10.3389/fonc.2021.621639] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Immune Checkpoint Inhibitors (ICI) have been progressively used in cancer treatment and produced unique toxicity profiles. This systematic review aims to comprehend the patterns and occurrence of treatment-related adverse events (trAEs) based on ICI. Methods PICOS/PRISMA methods were used to identify published English-language on PubMed, Web of Science, and Scopus from 2015 to 2020. Published clinical trials on ICI monotherapy, combined ICIs, and ICI plus other treatment with tabulated data on grade≥3 trAEs were included. Odds ratio (OR), χ2 tests were used to analyze for effect size and associations. Results This review included 145 clinical trials involving 21786 patients. Grade 3-5 trAEs were more common with ICI when they were plused with other treatments compared with ICI monotherapy(54.3% versus 17.7%, 46.1%, p<0.05). Grade 3-5 trAEs were also more common with CTLA-4 mAbs compared with anti-PD-1 and anti-PD-L1 (34.2% versus 15.1%, 13.6%, p<0.05). Hyperthyroidism (OR 3.8, 95%CI 1.7–8.6), nausea (OR 3.7, 95%CI 2.5–5.3), diarrhea (OR 2.7, 95%CI 2.2–3.2), colitis (OR 3.4, 95%CI 2.7–4.3), ALT increase (OR 4.9, 95%CI 3.9–6.1), AST increase (OR 3.8, 95%CI 3.0–4.9), pruritus (OR 2.4, 95%CI 1.5–3.9), rash (OR 2.8, 95%CI 2.1–3.8), fatigue (OR 2.8, 95%CI 2.2–3.7), decreased appetite (OR 2.4, 95%CI 1.5–3.8), and hypophysitis (OR 2.0, 95%CI 1.2–3.3) were more frequent with combined ICIs. Diarrhea (OR 8.1, 95%CI 6.4–10.3), colitis (OR 12.2, 95%CI 8.7–17.1), ALT increase (OR 5.1, 95%CI 3.5–7.4), AST increase (OR 4.2, 95%CI 2.8–6.3), pruritus (OR 4.1, 95%CI 2.0–8.4), rash (OR 4.4, 95%CI 2.9–6.8), hypophysitis (OR 12.1, 95%CI 6.3–23.4) were more common with CTLA-4 mAbs; whereas pneumonitis (OR 4.7, 95% CI 2.1–10.3) were more frequent with PD-1 mAbs. Conclusions Different immune checkpoint inhibitors are associated with different treatment-related adverse events profiles. A comprehensive data in this systematic review will provide comprehensive information for clinicians.
Collapse
Affiliation(s)
- Tao Ouyang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tao Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Liangliang Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
123
|
Tobin JWD, Bednarska K, Campbell A, Keane C. PD-1 and LAG-3 Checkpoint Blockade: Potential Avenues for Therapy in B-Cell Lymphoma. Cells 2021; 10:cells10051152. [PMID: 34068762 PMCID: PMC8151045 DOI: 10.3390/cells10051152] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
The dependence of cancer on an immunotolerant tumor microenvironment (TME) is well established. Immunotherapies that overcome tumor-induced immune suppression have been central to recent advancements in oncology. This is highlighted by the success of agents that interrupt PD-1 mediated immune suppression in a range of cancers. However, while PD-1 blockade has been paradigm-shifting in many malignancies, the majority of cancers show high rates of primary resistance to this approach. This has led to a rapid expansion in therapeutic targeting of other immune checkpoint molecules to provide combination immune checkpoint blockade (ICB), with one such promising approach is blockade of Lymphocyte Activation Gene 3 (LAG-3). Clinically, lymphoproliferative disorders show a wide spectrum of responses to ICB. Specific subtypes including classical Hodgkin lymphoma have demonstrated striking efficacy with anti-PD-1 therapy. Conversely, early trials of ICB have been relatively disappointing in common subtypes of Non-Hodgkin lymphoma. In this review, we describe the TME of common lymphoma subtypes with an emphasis on the role of prominent immune checkpoint molecules PD-1 and LAG3. We will also discuss current clinical evidence for ICB in lymphoma and highlight key areas for further investigation where synergistic dual checkpoint blockade of LAG-3 and PD-1 could be used to overcome ICB resistance.
Collapse
Affiliation(s)
- Joshua W. D. Tobin
- Mater Research Institute, University of Queensland, Brisbane, QLD 4102, Australia; (J.W.D.T.); (K.B.)
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia;
| | - Karolina Bednarska
- Mater Research Institute, University of Queensland, Brisbane, QLD 4102, Australia; (J.W.D.T.); (K.B.)
| | - Ashlea Campbell
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia;
| | - Colm Keane
- Mater Research Institute, University of Queensland, Brisbane, QLD 4102, Australia; (J.W.D.T.); (K.B.)
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia;
- Correspondence: ; Tel.: +617-3443-7912
| |
Collapse
|
124
|
Tsotridou E, Vasileiou E, Mantadakis E, Tragiannidis A. Safety and Efficacy of Immune Checkpoint Inhibitors in Children and Young Adults with Haematological Malignancies: Review and Future Perspectives. Cardiovasc Hematol Agents Med Chem 2021; 20:20-33. [PMID: 33970848 DOI: 10.2174/1871525719666210510171132] [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: 12/12/2020] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 11/22/2022]
Abstract
Despite the marked improvement in overall survival rates of paediatric patients with haematological malignancies that has been achieved during the last decades, there is still a pressing need for novel therapeutic approaches for the subset of patients with relapsed or refractory disease. Immune checkpoint inhibitors aim to induce potent anti-tumour immune responses by targeted blockade of inhibitory receptors and have shown great promise in preclinical models and studies in the adult population. However, paediatric malignancies present unique features and so far, experience with these agents remains limited. In the current review we present an overview of efficacy and safety data from case reports, case series and clinical trials employing the use of immune checkpoint inhibitors in children, adolescents and young adults with haematological malignancies. We also discuss new possibilities involving novel targets and combination treatments and provide a summary of the currently registered clinical trials.
Collapse
Affiliation(s)
- Eleni Tsotridou
- 2nd Department of Paediatrics, Childhood and Adolescent Hematology Oncology Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Eleni Vasileiou
- 2nd Department of Paediatrics, Childhood and Adolescent Hematology Oncology Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Elpis Mantadakis
- Department of Paediatrics, Paediatric Hematology/Oncology Unit, Democritus University of Thrace Faculty of Medicine, Alexandroupolis, Thrace, Greece
| | - Athanasios Tragiannidis
- 2nd Department of Paediatrics, Childhood and Adolescent Hematology Oncology Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| |
Collapse
|
125
|
Nieto Y, Gruschkus S, Valdez BC, Jones RB, Anderlini P, Hosing C, Popat U, Qazilbash M, Kebriaei P, Alousi A, Saini N, Srour S, Rezvani K, Ramdial J, Barnett M, Gulbis A, Shigle TL, Ahmed S, Iyer S, Lee H, Nair R, Parmar S, Steiner R, Dabaja B, Pinnix C, Gunther J, Cuglievan B, Mahadeo K, Khazal S, Chuang H, Champlin R, Shpall EJ, Andersson BS. Improved outcomes of high-risk relapsed Hodgkin lymphoma patients after high-dose chemotherapy: a 15-year analysis. Haematologica 2021; 107:899-908. [PMID: 33951890 PMCID: PMC8968895 DOI: 10.3324/haematol.2021.278311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Indexed: 02/03/2023] Open
Abstract
High-dose chemotherapy and autologous stem-cell transplant (HDC/ASCT) is standard treatment for chemosensitive relapsed classical Hodgkin lymphoma, although outcomes of high-risk relapse (HRR) patients remain suboptimal. We retrospectively analyzed all HRR classical Hodgkin lymphoma patients treated with HDC/ASCT at our institution between 01/01/2005 and 12/31/2019. HRR criteria included primary refractory disease/relapse within 1 year, extranodal extension, B symptoms, requiring more than one salvage line, or positron emission tomography (PET)-positive disease at ASCT. All patients met the same ASCT eligibility criteria. We treated 501 patients with BEAM (n=146), busulphan/melphalan (BuMel) (n=38), gemcitabine( Gem)/BuMel (n=189) and vorinostat/Gem/BuMel (n=128). The Gem/BuMel and vorinostat/Gem/BuMel cohorts had more HRR criteria and more patients with PET-positive disease at ASCT. Treatment with brentuximab vedotin (BV) or anti-PD1 prior to ASCT, PET-negative disease at ASCT, and maintenance BV increased over time. BEAM and BuMel predominated in earlier years (2005-2007), GemBuMel and BEAM in middle years (2008-2015), and vorinostat/GemBuMel and BEAM in later years (2016-2019). The median follow-up is 50 months (range, 6-186). Outcomes improved over time, with 2-year progressionfree survival (PFS)/overall survival (OS) rates of 58%/82% (2005-2007), 59%/83% (2008-2011), 71%/94% (2012-2015) and 86%/99% (2016- 2019) (P<0.0001). Five-year PFS/OS rates were 72%/87% after vorinostat/ GemBuMel, 55%/75% after GemBuMel, 45%/61% after BEAM, and 39%/57% after BuMel (PFS: P=0.0003; OS: P<0.0001). These differences persisted within the PET-negative and PET-positive subgroups. Prior BV and vorinostat/GemBuMel were independent predictors of more favorable outcome, whereas primary refractory disease, ≥2 salvage lines, bulky relapse, B symptoms and PET-positivity at ASCT correlated independently with unfavorable outcomes. In conclusion, post-HDC/ASCT outcomes of patients with HRR classic Hodgkin lymphoma have improved over the last 15 years. Pre-ASCT BV treatment and optimized synergistic HDC (vorinostat/GemBuMel) were associated with this improvement.
Collapse
Affiliation(s)
- Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center,YAGO NIETO
| | | | - Benigno C. Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Roy B. Jones
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Paolo Anderlini
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Muzaffar Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Neeraj Saini
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Melissa Barnett
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Alison Gulbis
- Pharmacy, University of Texas MD Anderson Cancer Center
| | | | - Sairah Ahmed
- Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center
| | - Swaminathan Iyer
- Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center
| | - Hun Lee
- Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center
| | - Ranjit Nair
- Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center
| | - Simrit Parmar
- Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center
| | - Raphael Steiner
- Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center
| | - Bouthaina Dabaja
- Radiation Oncology, University of Texas MD Anderson Cancer Center
| | - Chelsea Pinnix
- Radiation Oncology, University of Texas MD Anderson Cancer Center
| | - Jillian Gunther
- Radiation Oncology, University of Texas MD Anderson Cancer Center
| | | | - Kris Mahadeo
- Pediatrics, University of Texas MD Anderson Cancer Center
| | - Sajad Khazal
- Pediatrics, University of Texas MD Anderson Cancer Center
| | - Hubert Chuang
- Nuclear Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| | - Borje S. Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center
| |
Collapse
|
126
|
Huang RSP, Murugesan K, Montesion M, Pavlick DC, Mata DA, Hiemenz MC, Decker B, Frampton G, Albacker LA, Ross JS. Pan-cancer landscape of CD274 (PD-L1) copy number changes in 244 584 patient samples and the correlation with PD-L1 protein expression. J Immunother Cancer 2021; 9:e002680. [PMID: 33972391 PMCID: PMC8112409 DOI: 10.1136/jitc-2021-002680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Several studies have shown clinical outcomes data that support the use of CD274 (PD-L1) copy-number (CN) gains and/or losses as a biomarker for immune checkpoint inhibitor (ICPI). Here, we present the landscape of CD274 CN changes across a large cohort of solid tumor cases and correlate these with PD-L1 protein expression by immunohistochemistry. METHODS We analyzed all cases that underwent comprehensive genomic profiling (CGP) testing at Foundation Medicine between August 2014 and June 2020. CD274 CN changes were correlated with PD-L1 expression in tumor types where there were Food and Drug Administration approved companion diagnostic (CDx) claims and the CDx assay was used to assess PD-L1 expression. RESULTS In all, 244 584 samples representing 290 solid tumor types were included in the study. Overall, 17.6% (42 983/244 584) had CD274 CN gains (>specimen ploidy), 44.6% (108 970/244 584) were CD274 CN neutral, and 37.9% (92 631/244 584) had CD274 CN loss. Using different CN cut offs to define CD274 positivity resulted in different prevalence estimates: ploidy +1, 17.4% (42 636/244 584); ploidy +2, 6.2% (15 183/244 584); ploidy +3, 2.2% (5375/244 584); ploidy +4, 1.1% (2712/244 584); and ploidy +8, 0.2% (434/244 584). The prevalence of CN changes and CN positivity varied based on tumor type. CD274 CN gains were significantly associated with PD-L1 positivity in NSCLC, urothelial carcinoma, breast carcinoma, cervical carcinoma, esophagus squamous cell carcinoma (SCC) and head and neck SCC (ORs 3.3, 3.0, 2.0, 4.5. 3.8, 8.4, 1.4, respectively; p<0.05) and with microsatellite instability status in only clinically relevant tumor types (gastric adenocarcinoma, colorectal adenocarcinoma, uterine endometrial adenocarcinoma, esophageal adenocarcinoma and gastroesophageal junction adenocarcinoma (OR: 5.2, 1.9, 3.2, 3.7 and 6.5, respectively; p<0.05)). Conversely, CD274 CN changes were not significantly correlated with tumor mutational burden in almost all the tumor types. CONCLUSION CD274 CN changes and PD-L1 expression were highly correlated in multiple tumor types. These prevalence data on CD274 CN changes across a large cohort of different solid tumors can be used to design future clinical studies to assess whether CD274 CN changes could be a potential biomarker for ICPI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jeffrey S Ross
- Foundation Medicine Inc, Cambridge, Massachusetts, USA
- Department of Pathology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
127
|
Shi H, Yang Y. Identification of inhibitory immune checkpoints and relevant regulatory pathways in breast cancer stem cells. Cancer Med 2021; 10:3794-3807. [PMID: 33932112 PMCID: PMC8178503 DOI: 10.1002/cam4.3902] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint blockade (ICB) has become one of the most promising approaches to activating antitumor immunity. However, only a small subset of patients with breast cancer benefit from ICB treatment. To improve the therapeutic effect in the clinic, precision immunotherapy is proposed to accurately eliminate cancer stem cells that contribute to local recurrence or metastasis, but currently little is known about their immunological properties. In this study, breast cancer-specific datasets in The Cancer Genome Atlas were collected and analyzed by using multiple open-access web servers. We found that the immunophenotype of breast cancer was characterized by a hypoactive immune microenvironment and a low response to immune checkpoint blockade. The innate immune checkpoint CD200 and the adaptive immune checkpoint CD276, respectively, exhibited a strong correlation with basal/stem gene signature and invasiveness gene signature, both of which represent breast cancer stem cells. Wnt, TGF-β, and Hedgehog signaling, which are recognized as stemness-related pathways, showed a significant association with the expression of CD200 and CD276, suggesting cancer stem cell-specific immune checkpoints could be downregulated by inhibiting these pathways. Of note, levels of CD200 and CD276 expression were higher in TGF-β dominant breast cancer than in other immune types of breast cancer. We also identified gene signatures that represent Wnt, TGF-β, and Hedgehog signaling-related CD200 and CD276 expression in breast cancer stem cells. For the luminal A subtype, the patient group with a high level of these gene signatures plus a low infiltration of CD8+ T cells, or dendritic cells, or M1 macrophages had poor overall survival. Our study suggested that CD200 and CD276 are candidate inhibitory immune checkpoints in breast cancer stem cells, which are potentially regulated by Wnt, TGF-β, and Hedgehog signaling. Synergistic inhibition of these stemness-related pathways may improve the efficacy of ICB treatment targeting breast cancer stem cells in precision immunotherapy.
Collapse
Affiliation(s)
- Haojun Shi
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yisi Yang
- Graduate School of Asia-Pacific Studies, Waseda University, Tokyo, Japan
| |
Collapse
|
128
|
Olivares-Hernández A, Figuero-Pérez L, Terán-Brage E, López-Gutiérrez Á, Velasco ÁT, Sarmiento RG, Cruz-Hernández JJ, Miramontes-González JP. Resistance to Immune Checkpoint Inhibitors Secondary to Myeloid-Derived Suppressor Cells: A New Therapeutic Targeting of Haematological Malignancies. J Clin Med 2021; 10:jcm10091919. [PMID: 33925214 PMCID: PMC8124332 DOI: 10.3390/jcm10091919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/17/2021] [Accepted: 04/23/2021] [Indexed: 01/11/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a set of immature myeloid lineage cells that include macrophages, granulocytes, and dendritic cell precursors. This subpopulation has been described in relation to the tumour processes at different levels, including resistance to immunotherapy, such as immune checkpoint inhibitors (ICIs). Currently, multiple studies at the preclinical and clinical levels seek to use this cell population for the treatment of different haematological neoplasms, together with ICIs. This review addresses the different points in ongoing studies of MDSCs and ICIs in haematological malignancies and their future significance in routine clinical practice.
Collapse
Affiliation(s)
- Alejandro Olivares-Hernández
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Correspondence: (A.O.-H.); (J.P.M.-G.); Tel.: +34-923-29-11-00 (A.O.-H.); +34-983-42-04-00 (J.P.M.-G.); Fax: +34-923-29-13-25 (A.O.-H.); +34-983-21-53-65 (J.P.M.-G.)
| | - Luis Figuero-Pérez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Eduardo Terán-Brage
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Álvaro López-Gutiérrez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Álvaro Tamayo Velasco
- Department of Haematology, University Hospital of Valladolid, 47003 Valladolid, Spain;
| | - Rogelio González Sarmiento
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Juan Jesús Cruz-Hernández
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - José Pablo Miramontes-González
- Department of Internal Medicine, University Hospital Rio Hortega, 47012 Valladolid, Spain
- Department of Medicine, University of Valladolid, 45005 Valladolid, Spain
- Correspondence: (A.O.-H.); (J.P.M.-G.); Tel.: +34-923-29-11-00 (A.O.-H.); +34-983-42-04-00 (J.P.M.-G.); Fax: +34-923-29-13-25 (A.O.-H.); +34-983-21-53-65 (J.P.M.-G.)
| |
Collapse
|
129
|
Pezeshki PS, Eskian M, Hamblin MR, Rezaei N. Immune checkpoint inhibition in classical hodgkin lymphoma. Expert Rev Anticancer Ther 2021; 21:1003-1016. [PMID: 33857395 DOI: 10.1080/14737140.2021.1918548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Hodgkin lymphoma (HL) accounts for 10% of lymphoma cases every year. HL is often curable by conventional chemotherapy and radiotherapy. However, in case of relapsed or refractory HL (r/r HL) after autologous hematopoietic stem cell transplantation (ASCT), few treatment options are currently available. Blockade of the immune checkpoint receptors, programmed death receptor-1 (PD-1), or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expressed on T-cells, and their ligands expressed on tumor-associated antigen-presenting cells (APCs), and Hodgkin and Reed/Sternberg (HRS) cells can remove inhibitory signals from anti-tumor T cells. Checkpoint blockade using monoclonal antibodies could be a potential treatment. Nivolumab and pembrolizumab are approved antibodies for the treatment of r/r HL.Areas covered: This paper provides a comprehensive discussion of checkpoint inhibitors in HL treatment, including the most important clinical trials with mono- or combination therapies as a first or second-line treatment of HL.Expert opinion: Relatively high response rates and an acceptable safety profile of checkpoint inhibitors make them an effective therapy for HL. The combination of checkpoint inhibition with other conventional cancer treatments and identifying the mechanisms responsible for resistance to checkpoint inhibition may improve the efficacy and safety of this immunotherapy, and enhance patient quality of life.
Collapse
Affiliation(s)
- Parmida Sadat Pezeshki
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Eskian
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein South Africa
| | - Nima Rezaei
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran Iran
| |
Collapse
|
130
|
Human leukocyte antigen I is significantly downregulated in patients with myxoid liposarcomas. Cancer Immunol Immunother 2021; 70:3489-3499. [PMID: 33893830 PMCID: PMC8571150 DOI: 10.1007/s00262-021-02928-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/25/2021] [Indexed: 12/15/2022]
Abstract
The characteristics of the tumor immune microenvironment remains unclear in liposarcomas, and here we aimed to determine the prognostic impact of the tumor immune microenvironment across separate liposarcomas subtypes. A total of 70 liposarcoma patients with three subtypes: myxoid liposarcoma (n = 45), dedifferentiated liposarcoma (n = 17), and pleomorphic liposarcoma (n = 8) were enrolled. The presence of tumor infiltrating lymphocytes (CD4+ , CD8+ , FOXP3+ lymphocytes) and CD163+ macrophages and expression of HLA class I and PD-L1 were assessed by immunohistochemistry in the diagnostic samples; overall survival and progression-free survival were estimated from outcome data. For infiltrating lymphocytes and macrophages, dedifferentiated liposarcoma and pleomorphic liposarcoma patients had a significantly higher number than myxoid liposarcoma patients. While myxoid liposarcoma patients with a high number of macrophages were associated with worse overall and progression-free survival, dedifferentiated liposarcoma patients with high macrophage numbers showed a trend toward favorable prognosis. Expression of HLA class I was negative in 35 of 45 (77.8%) myxoid liposarcoma tumors, whereas all dedifferentiated liposarcoma and pleomorphic liposarcoma tumors expressed HLA class I. The subset of myxoid liposarcoma patients with high HLA class I expression had significantly poor overall and progression-free survival, while dedifferentiated liposarcoma patients with high HLA class I expression tended to have favorable outcomes. Only four of 17 (23.5%) dedifferentiated liposarcomas, two of eight (25%) pleomorphic liposarcomas, and no myxoid liposarcoma tumors expressed PD-L1. Our results demonstrate the unique immune microenvironment of myxoid liposarcomas compared to other subtypes of liposarcomas, suggesting that the approach for immunotherapy in liposarcomas should be based on subtype.
Collapse
|
131
|
Immunity reloaded: Deconstruction of the PD-1 axis in B cell lymphomas. Blood Rev 2021; 50:100832. [PMID: 33896649 DOI: 10.1016/j.blre.2021.100832] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/23/2022]
Abstract
Over the past decade therapies targeting the PD-1 axis with monoclonal antibodies to reinstate host immune function have revolutionized the clinical management of some cancers but have had minimal impact on others. This dichotomy is exemplified by B cell lymphomas. Whilst striking results are observed in classical Hodgkin Lymphoma (cHL) and Primary Mediastinal B Cell Lymphoma (PMBL), responses in other B cell lymphomas are infrequent. Even with cHL and PMBL, responses are not always durable and adverse effects can result in treatment discontinuation. A more nuanced approach to manipulate the PD-1 axis is required before the full benefits of PD-1 axis blockade can be realised. In this review, we provide an outline of PD-1 axis biology, including the range of cellular expression, the molecular mechanisms underlying regulation and the impacts of downstream signalling. These may permit the development of alternate strategies to PD-1 axis blockade to enhance the therapeutic efficacy in B cell lymphomas.
Collapse
|
132
|
Adenovirus-Mediated Inducible Expression of a PD-L1 Blocking Antibody in Combination with Macrophage Depletion Improves Survival in a Mouse Model of Peritoneal Carcinomatosis. Int J Mol Sci 2021; 22:ijms22084176. [PMID: 33920699 PMCID: PMC8073765 DOI: 10.3390/ijms22084176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated remarkable efficacy in a growing number of malignancies. However, overcoming primary or secondary resistances is difficult due to pharmacokinetics issues and side effects associated with high systemic exposure. Local or regional expression of monoclonal antibodies (mAbs) using gene therapy vectors can alleviate this problem. In this work, we describe a high-capacity adenoviral vector (HCA-EFZP-aPDL1) equipped with a mifepristone-inducible system for the controlled expression of an anti-programmed death ligand 1 (PD-L1) blocking antibody. The vector was tested in an immune-competent mouse model of colorectal cancer based on implantation of MC38 cells. A single local administration of HCA-EFZP-aPDL1 in subcutaneous lesions led to a significant reduction in tumor growth with minimal release of the antibody in the circulation. When the vector was tested in a more stringent setting (rapidly progressing peritoneal carcinomatosis), the antitumor effect was marginal even in combination with other immune-stimulatory agents such as polyinosinic-polycytidylic acid (pI:C), blocking mAbs for T cell immunoglobulin, mucin-domain containing-3 (TIM-3) or agonistic mAbs for 4-1BB (CD137). In contrast, macrophage depletion by clodronate liposomes enhanced the efficacy of HCA-EFZP-aPDL1. These results highlight the importance of addressing macrophage-associated immunoregulatory mechanisms to overcome resistance to ICIs in the context of colorectal cancer.
Collapse
|
133
|
Liu Y, Wang C, Li X, Dong L, Yang Q, Chen M, Shi F, Brock M, Liu M, Mei Q, Liu J, Nie J, Han W. Improved clinical outcome in a randomized phase II study of anti-PD-1 camrelizumab plus decitabine in relapsed/refractory Hodgkin lymphoma. J Immunother Cancer 2021; 9:jitc-2021-002347. [PMID: 33820822 PMCID: PMC8025784 DOI: 10.1136/jitc-2021-002347] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
Background Programmed death-1 (PD-1) blockade monotherapy induced durable remission in a subset of patients with relapsed/refractory classical Hodgkin lymphoma (cHL). We asked whether the anti-PD-1 agent, camrelizumab, combined with the DNA demethylating agent, decitabine, improves progression-free survival (PFS) in patients with relapsed/refractory cHL over camrelizumab alone. Methods This extended follow-up of an ongoing randomized phase II trial analyzed PFS among patients enrolled from January 2017 through July 2018. Sixty-one patients with relapsed/refractory cHL who were clinically naïve to PD-1 blockade and had received ≥2 previous therapies were randomized 1:2 to receive either camrelizumab (200 mg) monotherapy or camrelizumab (200 mg, day 8) combined with decitabine (10 mg/day, days 1–5) every 3 weeks. Results With a median follow-up of 34.5 months, complete remission was 79% (95% CI 63% to 90%) in the decitabine-plus-camrelizumab group versus 32% (95% CI 13% to 57%) in the camrelizumab group (p=0.001). Median duration of response was not reached in the decitabine-plus-camrelizumab group, with an estimated 63% (95% CI 46% to 75%) of patients maintaining a response at 24 months. Median PFS with decitabine-plus-camrelizumab therapy was 35.0 months (95% CI not reached) and 15.5 months (95% CI 8.4 to 22.7 months) with camrelizumab monotherapy (HR, 0.46; 95% CI 0.21 to 1.01; p=0.02). Female gender, lower tumor burden, and fewer previous therapies were favorable prognostic factors for durable remission with camrelizumab monotherapy. The PFS benefits of decitabine-plus-camrelizumab versus camrelizumab were observed in most subgroups, especially in patients with relative larger tumor burdens and those treated with ≥3 prior therapies. After decitabine-plus-camrelizumab treatment, the percentage increase of circulating peripheral central memory T-cells correlated with both improved clinical response and PFS, suggesting a putative biomarker of decitabine-plus-camrelizumab therapy for cHL. Conclusions Decitabine-plus-camrelizumab results in longer PFS compared with camrelizumab alone in patients with relapsed/refractory cHL. Trial registration numbers NCT02961101 and NCT03250962.
Collapse
Affiliation(s)
- Yang Liu
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Chunmeng Wang
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiang Li
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Liang Dong
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Qingming Yang
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Meixia Chen
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Fengxia Shi
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Malcolm Brock
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miao Liu
- Department of Statistics and Epidemiology, Chinese PLA General Hospital, Beijing, China
| | - Qian Mei
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jing Nie
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Weidong Han
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
134
|
Zhang W, Qiu Y, Xie X, Fu Y, Wang L, Cai Z. B7 Family Members in Lymphoma: Promising Novel Targets for Tumor Immunotherapy? Front Oncol 2021; 11:647526. [PMID: 33869045 PMCID: PMC8044412 DOI: 10.3389/fonc.2021.647526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
T cells play a vital role in the immune responses against tumors. Costimulatory or coinhibitory molecules regulate T cell activation. Immune checkpoint inhibitors, such as programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) have shown remarkable benefits in patients with various tumor, but few patients have displayed significant immune responses against tumors after PD-1/PD-L1 immunotherapy and many have been completely unresponsive. Thus, researchers must explore novel immune checkpoints that trigger durable antitumor responses and improve clinical outcomes. In this regard, other B7 family checkpoint molecules have been identified, namely PD-L2, B7-H2, B7-H3, B7-H4 and B7-H6. The aim of the present article was to address the expression, clinical significance and roles of B7 family molecules in lymphoma, as well as in T and NK cell-mediated tumor immunity. B7 family checkpoints may offer novel and immunotherapeutic strategies for patients with lymphoma.
Collapse
Affiliation(s)
- Wei Zhang
- School of Clinical Medicine, Binzhou Medical University, Yantai, China.,Central Laboratory, Linyi People's Hospital, Linyi, China
| | - Yu Qiu
- School of Clinical Medicine, Binzhou Medical University, Yantai, China.,Central Laboratory, Linyi People's Hospital, Linyi, China
| | - Xiaoli Xie
- Central Laboratory, Linyi People's Hospital, Linyi, China
| | - Yao Fu
- Central Laboratory, Linyi People's Hospital, Linyi, China
| | - Lijuan Wang
- School of Clinical Medicine, Binzhou Medical University, Yantai, China.,Central Laboratory, Linyi People's Hospital, Linyi, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
135
|
Li F, Chen Y, Pang M, Yang P, Jing H. Immune checkpoint inhibitors and cellular treatment for lymphoma immunotherapy. Clin Exp Immunol 2021; 205:1-11. [PMID: 33675535 DOI: 10.1111/cei.13592] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/30/2021] [Accepted: 02/21/2021] [Indexed: 11/29/2022] Open
Abstract
Malignant lymphoma (ML) is a common hematological malignancy with many subtypes. Patients with ML usually undergo traditional treatment failure and become relapsed or refractory (R/R) cases. Recently, immunotherapy, such as immune checkpoint inhibitors (ICIs) and cellular treatment, has gradually emerged and used in clinical trials with encouraging achievements for ML treatment, which exerts anti-tumor activity by blocking the immune evasion of tumor cells and enhancing the attack ability of immune cells. Targets of immune checkpoints include programmed cell death-1 (PD-1), programmed cell death-ligand 1 (PD-L1), cytotoxic T lymphocyte-associated protein 4 (CTLA-4), T cell immunoglobulin and ITIM domain (TIGIT), T cell immunoglobulin-3 (TIM-3) and lymphocyte activation gene 3 (LAG-3). Examples of cellular treatment are chimeric antigen receptor (CAR) T cells, cytokine-induced killer (CIK) cells and natural killer (NK) cells. This review aimed to present the current progress and future prospects of immunotherapy in lymphoma, with the focus upon ICIs and cellular treatment.
Collapse
Affiliation(s)
- F Li
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Y Chen
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - M Pang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - P Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - H Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| |
Collapse
|
136
|
Zam W, Ali L. Immune checkpoint inhibitors in the treatment of cancer. ACTA ACUST UNITED AC 2021; 17:103-113. [PMID: 33823768 DOI: 10.2174/1574884716666210325095022] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Immunotherapy drugs, known as immune checkpoint inhibitors (ICIs), work by blocking checkpoint proteins from binding with their partner proteins. The two main pathways that are specifically targeted in clinical practice are cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1) that showed potent immune-modulatory effects through their function as negative regulators of T cell activation. METHODS In view of the rapid and extensive development of this research field, we conducted a comprehensive review of the literature and update on the use of CTLA-4, PD-1 and PD-L1 targeted therapy in the treatment of several types of cancer including melanoma, non-small-cell lung carcinoma, breast cancer, hepatocellular carcinoma, hodgkin lymphoma, cervical cancer, head and neck squamous cell carcinoma. RESULTS Based on the last updated list released on March 2019, seven ICIs are approved by the FDA including ipilimumab, pembrolizumab, nivolumab, atezolizumab, avelumab, durvalumab, and cemiplimab. CONCLUSION This review also highlighted the most common adverse effects caused by ICIs and which affect people in different ways.
Collapse
Affiliation(s)
- Wissam Zam
- Department of Analytical and Food Chemistry, Faculty of Pharmacy, Al-Wadi International University, Homs. Syrian Arab Republic
| | - Lina Ali
- Department of Analytical and Food Chemistry, Faculty of Pharmacy, Tartous University, Tartous. Syrian Arab Republic
| |
Collapse
|
137
|
Csizmar CM, Ansell SM. Engaging the Innate and Adaptive Antitumor Immune Response in Lymphoma. Int J Mol Sci 2021; 22:3302. [PMID: 33804869 PMCID: PMC8038124 DOI: 10.3390/ijms22073302] [Citation(s) in RCA: 6] [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] [Received: 03/14/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has emerged as a powerful therapeutic strategy for many malignancies, including lymphoma. As in solid tumors, early clinical trials have revealed that immunotherapy is not equally efficacious across all lymphoma subtypes. For example, immune checkpoint inhibition has a higher overall response rate and leads to more durable outcomes in Hodgkin lymphomas compared to non-Hodgkin lymphomas. These observations, combined with a growing understanding of tumor biology, have implicated the tumor microenvironment as a major determinant of treatment response and prognosis. Interactions between lymphoma cells and their microenvironment facilitate several mechanisms that impair the antitumor immune response, including loss of major histocompatibility complexes, expression of immunosuppressive ligands, secretion of immunosuppressive cytokines, and the recruitment, expansion, and skewing of suppressive cell populations. Accordingly, treatments to overcome these barriers are being rapidly developed and translated into clinical trials. This review will discuss the mechanisms of immune evasion, current avenues for optimizing the antitumor immune response, clinical successes and failures of lymphoma immunotherapy, and outstanding hurdles that remain to be addressed.
Collapse
Affiliation(s)
| | - Stephen M. Ansell
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
138
|
Andrade-Gonzalez X, Ansell SM. Novel Therapies in the Treatment of Hodgkin Lymphoma. Curr Treat Options Oncol 2021; 22:42. [PMID: 33755826 DOI: 10.1007/s11864-021-00840-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2021] [Indexed: 12/22/2022]
Abstract
OPINION STATEMENT Patients with Hodgkin lymphoma (HL) can achieve excellent response and survival rates following frontline combination chemo- and radiation therapy. However, about 10-15% of patients will experience disease relapse which is associated with poor outcomes. Recent breakthroughs in understanding the mechanisms of oncogenicity and interactions within the tumor microenvironment have resulted in development of novel drugs for treatment of patients with HL. Utilizing this information, treatment of newly diagnosed and relapsed HL has become a rapidly evolving field with multiple clinical trials evaluating novel treatment approaches incorporating targeted immunotherapy. In the frontline setting, the use of novel drugs may allow for de-escalation of therapy to avoid long-term complications associated with bleomycin and consolidation radiation therapy. Patients with early-stage, non-bulky disease are candidates for omitting radiation therapy using treatment combinations that include upfront use of brentuximab vedotin or nivolumab. In patients with advanced disease, the addition of brentuximab vedotin to a chemotherapy backbone is currently the standard of care in our practice, particularly in patients with a contraindication for receiving bleomycin. Future investigations in patients with advanced-stage HL will focus on establishing a new standard of care by comparing brentuximab vedotin and nivolumab in combination with chemotherapy (BV-AVD vs. N-AVD) and decreasing the risk of relapse by exploring consolidation therapy in patients with high-risk disease. In patients who have relapsed or are refractory to first-line therapy, salvage treatment has incorporated brentuximab vedotin or PD-1 checkpoint inhibitors to improve response rates of cytotoxic chemotherapy thereby improving the probability of a successful stem cell transplant. Post-transplant consolidation with brentuximab is currently standard of care in patients with high-risk disease. Patients who relapse following autologous stem cell transplant now have an expanded armamentarium of chemo- and immunotherapy options. However, the challenge is to determine the sequence of therapy after prior brentuximab or checkpoint inhibitor exposure.
Collapse
Affiliation(s)
| | - Stephen M Ansell
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
139
|
Ferrari C, Maggialetti N, Masi T, Nappi AG, Santo G, Niccoli Asabella A, Rubini G. Early Evaluation of Immunotherapy Response in Lymphoma Patients by 18F-FDG PET/CT: A Literature Overview. J Pers Med 2021; 11:217. [PMID: 33803667 PMCID: PMC8002936 DOI: 10.3390/jpm11030217] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy is a promising therapeutic strategy both for solid and hematologic tumors, such as in Hodgkin (HL) and non-Hodgkin lymphoma (NHL). In particular, immune-checkpoint inhibitors, such as nivolumab and pembrolizumab, are increasingly used for the treatment of refractory/relapsed HL. At the same time, evidence of chimeric antigen receptor (CAR)-T-cell immunotherapy efficacy mostly in NHL is growing. In this setting, the challenge is to identify an appropriate imaging method to evaluate immunotherapy response. The role of 18F-Fluorodeoxyglucose (18F-FDG) positron-emission tomography/computed tomography (PET/CT), especially in early evaluation, is under investigation in order to guide therapeutic strategies, taking into account the possible atypical responses (hyperprogression and pseudoprogression) and immune-related adverse events that could appear on PET images. Herein, we aimed to present a critical overview about the role of 18F-FDG PET/CT in evaluating treatment response to immunotherapy in lymphoma patients.
Collapse
Affiliation(s)
- Cristina Ferrari
- Section of Nuclear Medicine, DIM, University Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy; (T.M.); (A.G.N.); (G.S.); (G.R.)
| | - Nicola Maggialetti
- Section of Radiodiagnostic, DSMBNOS, University Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Tamara Masi
- Section of Nuclear Medicine, DIM, University Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy; (T.M.); (A.G.N.); (G.S.); (G.R.)
| | - Anna Giulia Nappi
- Section of Nuclear Medicine, DIM, University Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy; (T.M.); (A.G.N.); (G.S.); (G.R.)
| | - Giulia Santo
- Section of Nuclear Medicine, DIM, University Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy; (T.M.); (A.G.N.); (G.S.); (G.R.)
| | | | - Giuseppe Rubini
- Section of Nuclear Medicine, DIM, University Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy; (T.M.); (A.G.N.); (G.S.); (G.R.)
| |
Collapse
|
140
|
Kuruvilla J, Ramchandren R, Santoro A, Paszkiewicz-Kozik E, Gasiorowski R, Johnson NA, Fogliatto LM, Goncalves I, de Oliveira JSR, Buccheri V, Perini GF, Goldschmidt N, Kriachok I, Dickinson M, Komarnicki M, McDonald A, Ozcan M, Sekiguchi N, Zhu Y, Nahar A, Marinello P, Zinzani PL. Pembrolizumab versus brentuximab vedotin in relapsed or refractory classical Hodgkin lymphoma (KEYNOTE-204): an interim analysis of a multicentre, randomised, open-label, phase 3 study. Lancet Oncol 2021; 22:512-524. [PMID: 33721562 DOI: 10.1016/s1470-2045(21)00005-x] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND PD-1 blockade via pembrolizumab monotherapy has shown antitumour activity and toxicity in patients with relapsed or refractory classical Hodgkin lymphoma. Here, we present interim analyses from the KEYNOTE-204 study evaluating pembrolizumab versus brentuximab vedotin for relapsed or refractory classical Hodgkin lymphoma. METHODS In this randomised, open-label, phase 3 study, patients aged 18 years or older with relapsed or refractory classical Hodgkin lymphoma with measurable disease and an Eastern Cooperative Oncology Group performance status of 0 or 1 who were ineligible for or had relapsed after autologous haematopoietic stem-cell transplantation (HSCT) were enrolled at 78 hospitals and cancer centres in 20 countries and territories. Patients were randomly assigned (1:1) with an interactive voice response system to pembrolizumab 200 mg intravenously every 3 weeks or brentuximab vedotin 1·8 mg/kg intravenously every 3 weeks. Randomisation was stratified by previous autologous HSCT and status after front-line therapy. Results from the second interim analysis are presented here, with a database cutoff of Jan 16, 2020. The dual primary endpoints assessed in the intention-to-treat population were progression-free survival as assessed by blinded independent central review, and overall survival (not analysed at this interim analysis). Safety was assessed in all patients who received at least one dose of the study drug. This study is registered with ClinicalTrials.gov, NCT02684292. Recruitment for this trial is closed. FINDINGS Between July 8, 2016, and July 13, 2018, 151 patients were randomly assigned to pembrolizumab and 153 to brentuximab vedotin. After a median time from randomisation to data cutoff of 25·7 months (IQR 23·4-33·0), median progression-free survival was 13·2 months (95% CI 10·9-19·4) for pembrolizumab versus 8·3 months (5·7-8·8) for brentuximab vedotin (hazard ratio 0·65 [95% CI 0·48-0·88]; p=0·0027). The most common grade 3-5 treatment-related adverse events were pneumonitis (six [4%] of 148 patients in the pembrolizumab group vs one [1%] of 152 patients in the brentuximab vedotin group), neutropenia (three [2%] vs 11 [7%]), decreased neutrophil count (one [1%] vs seven [5%]), and peripheral neuropathy (one [1%] vs five [3%]). Serious treatment-related adverse events occurred in 24 (16%) of 148 patients receiving pembrolizumab and 16 (11%) of 152 patients receiving brentuximab vedotin. One treatment-related death due to pneumonia occurred in the pembrolizumab group. INTERPRETATION Pembrolizumab showed statistically significant and clinically meaningful improvement in progression-free survival compared with brentuximab vedotin, with safety consistent with previous reports. These data support pembrolizumab as the preferred treatment option for patients with relapsed or refractory classical Hodgkin lymphoma who have relapsed post-autologous HSCT or are ineligible for autologous HSCT. FUNDING Merck Sharp & Dohme Corp (a subsidiary of Merck & Co, Inc, Kenilworth, NJ, USA).
Collapse
Affiliation(s)
| | | | - Armando Santoro
- Humanitas University, Department of Biomedical Sciences, Milan, Italy
| | | | | | | | | | - Iara Goncalves
- Fundação Pio XII-Hospital de Câncer de Barretos, São Paulo, Brazil
| | | | - Valeria Buccheri
- Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | - Michael Dickinson
- Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | | | | | - Muhit Ozcan
- Ankara University School of Medicine, Ankara, Turkey
| | - Naohiro Sekiguchi
- Department of Hematology, National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Ying Zhu
- Concord Hospital, Merck & Co, Kenilworth, NJ, USA
| | - Akash Nahar
- Concord Hospital, Merck & Co, Kenilworth, NJ, USA
| | | | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | | |
Collapse
|
141
|
Spatial signatures identify immune escape via PD-1 as a defining feature of T-cell/histiocyte-rich large B-cell lymphoma. Blood 2021; 137:1353-1364. [PMID: 32871584 PMCID: PMC8555417 DOI: 10.1182/blood.2020006464] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/18/2020] [Indexed: 01/28/2023] Open
Abstract
T-cell/histiocyte-rich large B-cell lymphoma (TCRLBCL) is an aggressive variant of diffuse large B-cell lymphoma (DLBCL) characterized by rare malignant B cells within a robust but ineffective immune cell infiltrate. The mechanistic basis of immune escape in TCRLBCL is poorly defined and not targeted therapeutically. We performed a genetic and quantitative spatial analysis of the PD-1/PD-L1 pathway in a multi-institutional cohort of TCRLBCLs and found that malignant B cells harbored PD-L1/PD-L2 copy gain or amplification in 64% of cases, which was associated with increased PD-L1 expression (P = .0111). By directed and unsupervised spatial analyses of multiparametric cell phenotypic data within the tumor microenvironment, we found that TCRLBCL is characterized by tumor-immune "neighborhoods" in which malignant B cells are surrounded by exceptionally high numbers of PD-L1-expressing TAMs and PD-1+ T cells. Furthermore, unbiased clustering of spatially resolved immune signatures distinguished TCRLBCL from related subtypes of B-cell lymphoma, including classic Hodgkin lymphoma (cHL) and DLBCL-NOS. Finally, we observed clinical responses to PD-1 blockade in 3 of 5 patients with relapsed/refractory TCRLBCL who were enrolled in clinical trials for refractory hematologic malignancies (NCT03316573; NCT01953692), including 2 complete responses and 1 partial response. Taken together, these data implicate PD-1 signaling as an immune escape pathway in TCRLBCL and also support the potential utility of spatially resolved immune signatures to aid the diagnostic classification and immunotherapeutic prioritization of diverse tumor types.
Collapse
|
142
|
Allen PB, Savas H, Evens AM, Advani RH, Palmer B, Pro B, Karmali R, Mou E, Bearden J, Dillehay G, Bayer RA, Eisner RM, Chmiel JS, O'Shea K, Gordon LI, Winter JN. Pembrolizumab followed by AVD in untreated early unfavorable and advanced-stage classical Hodgkin lymphoma. Blood 2021; 137:1318-1326. [PMID: 32992341 PMCID: PMC7955404 DOI: 10.1182/blood.2020007400] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/20/2020] [Indexed: 02/06/2023] Open
Abstract
Pembrolizumab, a humanized IgG4 monoclonal antibody targeting programmed death-1 protein, has demonstrated efficacy in relapsed/refractory classical Hodgkin lymphoma (cHL). To assess the complete metabolic response (CMR) rate and safety of pembrolizumab monotherapy in newly diagnosed cHL, we conducted a multicenter, single-arm, phase 2 investigator-initiated trial of sequential pembrolizumab and doxorubicin, vinblastine, and dacarbazine (AVD) chemotherapy. Patients ≥18 years of age with untreated, early, unfavorable, or advanced-stage disease were eligible for treatment. Thirty patients (early unfavorable stage, n = 12; advanced stage, n = 18) were treated with 3 cycles of pembrolizumab monotherapy followed by AVD for 4 to 6 cycles, depending on stage and bulk. Twelve had either large mediastinal masses or bulky disease (>10 cm). After pembrolizumab monotherapy, 11 patients (37%) demonstrated CMRs, and an additional 7 of 28 (25%) patients with quantifiable positron emission tomography computed tomography scans had >90% reduction in metabolic tumor volume. All patients achieved CMR after 2 cycles of AVD and maintained their responses at the end of treatment. With a median follow-up of 22.5 months (range, 14.2-30.6) there were no changes in therapy, progressions, or deaths. No patients received consolidation radiotherapy, including those with bulky disease. Therapy was well tolerated. The most common immune-related adverse events were grade 1 rash (n = 6) and grade 2 infusion reactions (n = 4). One patient had reversible grade 4 transaminitis and a second had reversible Bell's palsy. Brief pembrolizumab monotherapy followed by AVD was both highly effective and safe in patients with newly diagnosed cHL, including those with bulky disease. This trial was registered at www.clinicaltrials.gov as #NCT03226249.
Collapse
Affiliation(s)
| | - Hatice Savas
- Department of Radiology and
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Andrew M Evens
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | | | - Brett Palmer
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Barbara Pro
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Reem Karmali
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Eric Mou
- Stanford Cancer Institute, Palo Alto, CA; and
| | - Jeffrey Bearden
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Gary Dillehay
- Department of Radiology and
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Robert A Bayer
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Robert M Eisner
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Joan S Chmiel
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Kaitlyn O'Shea
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Leo I Gordon
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jane N Winter
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
143
|
Kaloyannidis P, Al Shaibani E, Moinnudin A, Al Anezi K, Al Hashmi H. Repeated courses of escalating doses of Nivolumab in refractory Hodgkin lymphoma with recurrent relapses post allografting: A safe and effective treatment approach. Hematol Rep 2021; 13:8780. [PMID: 33747411 PMCID: PMC7967268 DOI: 10.4081/hr.2021.8780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/01/2021] [Indexed: 01/01/2023] Open
Abstract
For patients with Hodgkin Lymphoma (HL) who experience relapse post allogeneic stem cell transplantation, limited treatment options exist, and the ultimate outcome is poor. Recently, the programmed cell death protein-1 (PD-1) inhibitors have shown remarkable efficacy in patients with refractory/relapsed HL, also demonstrating an acceptable safety profile. However, due to effects on T-cell activity, the use of PD-1 inhibitors post allografting may potentially increase the risk of treatment-emergent graft versus host disease. We herein report the clinical course of a patient who experienced multiple relapses of HL post allogeneic stem cell transplantation. He failed several treatment modalities but he responded to escalating doses of the PD-1 inhibitor nivolumab, given at two different treatment time points, also demonstrating minimal and easily manageable toxicity.
Collapse
Affiliation(s)
| | | | - Asif Moinnudin
- Departments of Medical Imaging, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Khalid Al Anezi
- Departments of Adult Hematology and Stem Cell Transplantation
| | - Hani Al Hashmi
- Departments of Adult Hematology and Stem Cell Transplantation
| |
Collapse
|
144
|
Wang C, Liu Y, Dong L, Li X, Yang Q, Brock MV, Mei Q, Liu J, Chen M, Shi F, Liu M, Nie J, Han W. Efficacy of Decitabine plus Anti-PD-1 Camrelizumab in Patients with Hodgkin Lymphoma Who Progressed or Relapsed after PD-1 Blockade Monotherapy. Clin Cancer Res 2021; 27:2782-2791. [PMID: 33674274 DOI: 10.1158/1078-0432.ccr-21-0133] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/17/2021] [Accepted: 03/02/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Programmed death-1 (PD-1) blockade monotherapy is effective in relapsed/refractory classical Hodgkin lymphoma (cHL), but a subset of patients is recalcitrant to PD-1 inhibitors and only a minority of patients achieves durable remission. Effective treatment regimens for those with relapsed/progressive cHL after single-agent anti-PD-1 are urgently needed. Anti-PD-1 combination with the DNA-demethylating agent decitabine showed positive preliminary results in our test cohort patients who were resistant to anti-PD-1. Here, we assess the efficacy of decitabine plus anti-PD-1 therapy in an expansion cohort and after longer follow-up. PATIENTS AND METHODS We present the response and progression-free survival rates from patients with relapsed/refractory cHL who relapsed/progressed after prior anti-PD-1 monotherapy, and who received decitabine (10 mg/day, days 1-5) plus the anti-PD-1 camrelizumab (200 mg, day 8), every 3 weeks in a phase II trial (ClinicalTrials.gov: NCT02961101 and NCT03250962). RESULTS Overall, 51 patients (test cohort: 25, expansion cohort: 26) were treated and 50 evaluated for efficacy. The objective response rate was 52% [nine complete responses (CR); 36%] in the test cohort, and 68% (six CRs; 24%) in the expansion cohort. Median progression-free survival with decitabine plus camrelizumab was 20.0 and 21.6 months, respectively, which was significantly longer than that achieved with prior anti-PD-1 monotherapy. Durable response was observed in an estimated 78% of patients who achieved CR at 24 months. After decitabine plus camrelizumab, the ratio increase of circulating peripheral central memory T cells directly correlated with both clinical response and progression-free survival. CONCLUSIONS Decitabine plus camrelizumab is associated with high response rates and long-term benefits in patients with relapsed/refractory cHL who failed PD-1 inhibitors.
Collapse
Affiliation(s)
- Chunmeng Wang
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Yang Liu
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Liang Dong
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Xiang Li
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Qingming Yang
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Malcolm V Brock
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Qian Mei
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Jiejie Liu
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Meixia Chen
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Fengxia Shi
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China
| | - Miao Liu
- Department of Statistics and Epidemiology, Graduate School of Chinese PLA General Hospital, Beijing, P.R. China
| | - Jing Nie
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China.
| | - Weidong Han
- Department of Bio-therapeutic, The First Medical Centre in Chinese PLA General Hospital, Beijing, P.R. China.
| |
Collapse
|
145
|
Gamboa-Cedeño AM, Díaz M, Cristaldo N, Otero V, Schutz N, Fantl D, Cugliari S, Zerga M, Rojas-Bilbao E, Jauk F, García Rivello H, Nuñez M, Ranuncolo SM. Apoptotic regulator BCL-2 blockade as a potential therapy in classical Hodgkin Lymphoma. Life Sci 2021; 268:118979. [PMID: 33421528 DOI: 10.1016/j.lfs.2020.118979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/22/2022]
Abstract
The challenge in classical Hodgkin Lymphoma (cHL) management is the 30-40% of refractory/relapsed cases. AIMS The aim of this work was to determine whether NIK and BCL-2 could be useful as prognosis biomarkers in cHL. In addition, we evaluated BCL-2 as a directed-therapy in cHL cell lines using venetoclax. MAIN METHODS We evaluated NIK and BCL-2 expression in 112 untreated cHL patients' lymph-node biopsies by immunohistochemistry. cHL cell lines were treated with venetoclax alone or combined with vincristine or doxorubicin. Cell viability, metabolic activity and cell death were analyzed by trypan-blue exclusion method, MTS assay and FDA/IP staining respectively. KEY FINDINGS No correlation between NIK or BCL-2 expression and the majority of the clinical parameters was found. Patients with ≥60% BCL-2+ HRS-cells had a shorter disease-free survival (DFS) and overall survival (OS) (p = 0.002, p = 0.02 respectively). A decision tree analysis, in a 30 patients subgroup, showed that patients with <60% NIK+ HRS-cells but with ≥60% BCL-2+ HRS-cells had a worse outcome in terms of DFS and OS. These parameters performed better as prognosis indicators as compared to the diagnosis bone marrow status. Human cHL cell lines U-H01, KM-H2, L1236, SUPHD1, L540 showed sensitivity to venetoclax. The co-treatment effect of venetoclax and vincristine or doxorubicin on cell viability was diverse depending on the cell line evaluated. SIGNIFICANCE BCL-2 should be considered as a prognosis biomarker as well as a potential new therapeutic target in cHL. We report for the first time the cytotoxic effect of venetoclax in human cHL cell lines.
Collapse
Affiliation(s)
| | - Mariángeles Díaz
- Research Area, Institute of Oncology "A.H. Roffo", School of Medicine (FMED), Universidad de Buenos Aires, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Argentina
| | - Nancy Cristaldo
- Hematology, Italian Hospital of Buenos Aires (HIBA), Buenos Aires, Argentina
| | - Victoria Otero
- Hematology, Italian Hospital of Buenos Aires (HIBA), Buenos Aires, Argentina
| | - Natalia Schutz
- Hematology, Italian Hospital of Buenos Aires (HIBA), Buenos Aires, Argentina
| | - Dorotea Fantl
- Hematology, Italian Hospital of Buenos Aires (HIBA), Buenos Aires, Argentina
| | - Silvana Cugliari
- Hematology, Institute of Oncology "A.H. Roffo", School of Medicine (FMED), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marta Zerga
- Hematology, Institute of Oncology "A.H. Roffo", School of Medicine (FMED), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Erica Rojas-Bilbao
- Pathology, Institute of Oncology "A.H. Roffo", School of Medicine (FMED), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico Jauk
- Sequencing Laboratory, Italian Hospital of Buenos Aires (HIBA), Buenos Aires, Argentina
| | | | - Myriam Nuñez
- School of Pharmacy and Biochemistry (FFyB), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Stella Maris Ranuncolo
- Traslational Medicine and Biomedical Engineering Institute (IMTIB), Buenos Aires, Argentina; Research Area, Institute of Oncology "A.H. Roffo", School of Medicine (FMED), Universidad de Buenos Aires, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Argentina.
| |
Collapse
|
146
|
Swoboda R, Giebel S, Knopińska-Posłuszny W, Chmielowska E, Drozd-Sokołowska J, Paszkiewicz-Kozik E, Kulikowski W, Taszner M, Mendrek W, Najda J, Czerw T, Olszewska-Szopa M, Czyż A, Giza A, Spychałowicz W, Subocz E, Szwedyk P, Krzywon A, Wilk A, Zaucha JM. High efficacy of BGD (bendamustine, gemcitabine, and dexamethasone) in relapsed/refractory Hodgkin Lymphoma. Ann Hematol 2021; 100:1755-1767. [PMID: 33625572 PMCID: PMC8195914 DOI: 10.1007/s00277-021-04448-5] [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: 05/30/2020] [Accepted: 02/02/2021] [Indexed: 10/25/2022]
Abstract
The optimal salvage therapy in relapsed/refractory Hodgkin lymphoma (R/R HL) has not been defined so far. The goal of this multicenter retrospective study was to evaluate efficacy and safety of BGD (bendamustine, gemcitabine, dexamethasone) as a second or subsequent line of therapy in classical R/R HL. We have evaluated 92 consecutive R/R HL patients treated with BGD. Median age was 34.5 (19-82) years. Fifty-eight patients (63%) had received 2 or more lines of chemotherapy, 32 patients (34.8%) radiotherapy, and 21 patients (22.8%) an autologous hematopoietic stem cell transplantation (autoHCT). Forty-four patients (47.8%) were resistant to first line of chemotherapy. BGD therapy consisted of bendamustine 90 mg/m2 on days 1 and 2, gemcitabine 800 mg/m2 on days 1 and 4, dexamethasone 40 mg on days 1-4. Median number of BGD cycles was 4 (2-7). The following adverse events ≥ 3 grade were noted: neutropenia (22.8%), thrombocytopenia (20.7%), anemia (15.2%), infections (10.9%), AST/ALT increase (2.2%), and skin rush (1.1%). After BGD therapy, 51 (55.4%) patients achieved complete remission, 23 (25%)-partial response, 7 (7.6%)-stable disease, and 11 (12%) patients experienced progression disease. AutoHCT was conducted in 42 (45.7%) patients after BGD therapy, and allogeneic HCT (alloHCT) in 16 (17.4%) patients. Median progression-free survival was 21 months. BGD is a highly effective, well-tolerated salvage regimen for patients with R/R HL, providing an excellent bridge to auto- or alloHCT.
Collapse
Affiliation(s)
- Ryszard Swoboda
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice branch, Gliwice, Poland
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice branch, Gliwice, Poland
| | | | - Ewa Chmielowska
- Oncologic Hospital, Tomaszów Mazowiecki, Poland.,Department of Oncology, Oncology Centre, Bydgoszcz, Poland
| | - Joanna Drozd-Sokołowska
- Department of Hematology, Oncology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Paszkiewicz-Kozik
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw branch, Warsaw, Poland
| | - Waldemar Kulikowski
- Department of Hematology, Independent Public Health Care Ministry of the Interior of Warmia and Mazury Oncology Center, Olsztyn, Poland
| | - Michał Taszner
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Włodzimierz Mendrek
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice branch, Gliwice, Poland
| | - Jacek Najda
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice branch, Gliwice, Poland
| | - Tomasz Czerw
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice branch, Gliwice, Poland
| | - Magdalena Olszewska-Szopa
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wrocław Medical University, Wrocław, Poland
| | - Anna Czyż
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wrocław Medical University, Wrocław, Poland
| | - Agnieszka Giza
- Department of Hematology, Jagiellonian University, Krakow, Poland
| | - Wojciech Spychałowicz
- Internal Medicine and Oncology Clinic, Silesian Medical University, Katowice, Poland
| | - Edyta Subocz
- Department of Hematology, Military Institute of Medicine, Warsaw, Poland
| | - Paweł Szwedyk
- Department of Hematology, Ludwik Rydygier Hospital, Krakow, Poland
| | - Aleksandra Krzywon
- Department of Biostatistics and Bioinformatics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Agata Wilk
- Department of Biostatistics and Bioinformatics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Jan Maciej Zaucha
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
147
|
Borchmann P, Plütschow A, Kobe C, Greil R, Meissner J, Topp MS, Ostermann H, Dierlamm J, Mohm J, Thiemer J, Sökler M, Kerkhoff A, Ahlborn M, Halbsguth TV, Martin S, Keller U, Balabanov S, Pabst T, Vogelhuber M, Hüttmann A, Wilhelm M, Zijlstra JM, Moccia A, Kuhnert G, Bröckelmann PJ, von Tresckow B, Fuchs M, Klimm B, Rosenwald A, Eich H, Baues C, Marnitz S, Hallek M, Diehl V, Dietlein M, Engert A. PET-guided omission of radiotherapy in early-stage unfavourable Hodgkin lymphoma (GHSG HD17): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2021; 22:223-234. [PMID: 33539742 DOI: 10.1016/s1470-2045(20)30601-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Combined-modality treatment consisting of chemotherapy and consolidation radiotherapy is standard of care for patients with early-stage unfavourable Hodgkin lymphoma. However, the use of radiotherapy can have long-term sequelae, which is of particular concern, as Hodgkin lymphoma is frequently diagnosed in young adults with a median age of approximately 30 years. In the German Hodgkin Study Group HD17 trial, we investigated whether radiotherapy can be omitted without loss of efficacy in patients who have a complete metabolic response after receiving two cycles of escalated doses of etoposide, cyclophosphamide, and doxorubicin, and regular doses of bleomycin, vincristine, procarbazine, and prednisone (eBEACOPP) plus two cycles of doxorubicin, bleomycin, vinblastine, dacarbazine (ABVD) chemotherapy (2 + 2). METHODS In this multicentre, open-label, randomised, phase 3 trial, patients (aged 18-60 years) with newly diagnosed early-stage unfavourable Hodgkin lymphoma (all histologies) and an Eastern Cooperative Oncology Group performance status of 2 or less were enrolled at 224 hospitals and private practices in Germany, Switzerland, Austria, and the Netherlands. Patients were randomly assigned (1:1) to receive either standard combined-modality treatment, consisting of the 2 + 2 regimen (eBEACOPP consisted of 1250 mg/m2 intravenous cyclophosphamide on day 1, 35 mg/m2 intravenous doxorubicin on day 1, 200 mg/m2 intravenous etoposide on days 1-3, 100 mg/m2 oral procarbazine on days 1-7, 40 mg/m2 oral prednisone on days 1-14, 1·4 mg/m2 intravenous vincristine on day 8 [maximum dose of 2 mg per cycle], and 10 mg/m2 intravenous bleomycin on day 8; ABVD consisted of 25 mg/m2 intravenous doxorubicin, 10 mg/m2 intravenous bleomycin, 6 mg/m2 intravenous vinblastine, and 375 mg/m2 intravenous dacarbazine, all given on days 1 and 15) followed by 30 Gy involved-field radiotherapy (standard combined-modality treatment group) or PET4-guided treatment, consisting of the 2 + 2 regimen followed by 30 Gy of involved-node radiotherapy only in patients with positive PET at the end of four cycles of chemotherapy (PET4; PET4-guided treatment group). Randomisation was done centrally and used the minimisation method and seven stratification factors (centre, age, sex, clinical symptoms, disease localisation, albumin concentration, and bulky disease), and patients and investigators were masked to treatment allocation until central review of the PET4 examination had been completed. With the final analysis presented here, the primary objective was to show non-inferiority of the PET4-guided strategy in a per-protocol analysis of the primary endpoint of progression-free survival. We defined non-inferiority as an absolute difference of 8% in the 5-year progression-free survival estimates between the two groups. Safety analyses were done in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, NCT01356680. FINDINGS Between Jan 13, 2012, and March 21, 2017, we enrolled and randomly assigned 1100 patients to the standard combined-modality treatment group (n=548) or to the PET4-guided treatment group (n=552); two patients in each group were found ineligible after randomisation. At a median follow-up of 46·2 months (IQR 32·7-61·2), 5-year progression-free survival was 97·3% (95% CI 94·5-98·7) in the standard combined-modality treatment group and 95·1% (92·0-97·0) in the PET4-guided treatment group (hazard ratio 0·523 [95% CI 0·226-1·211]). The between-group difference was 2·2% (95% CI -0·9 to 5·3) and excluded the non-inferiority margin of 8%. The most common grade 3 or 4 acute haematological adverse events were leucopenia (436 [83%] of 528 patients in the standard combined-modality treatment group vs 443 [84%] of 529 patients in the PET4-guided treatment group) and thrombocytopenia (139 [26%] vs 176 [33%]), and the most frequent acute non-haematological toxic effects were infection (32 [6%] vs 40 [8%]) and nausea or vomiting (38 [7%] vs 29 [6%]). The most common acute radiotherapy-associated adverse events were dysphagia (26 [6%] in the standard combined-modality treatment group vs three [2%] in the PET4-guided treatment group) and mucositis (nine [2%] vs none). 229 serious adverse events were reported by 161 (29%) of 546 patients in the combined-modality treatment group, and 235 serious adverse events were reported by 164 (30%) of 550 patients in the PET4-guided treatment group. One suspected unexpected serious adverse reaction (infection) leading to death was reported in the PET4-guided treatment group. INTERPRETATION PET4-negativity after treatment with 2 + 2 chemotherapy in patients with newly diagnosed early-stage unfavourable Hodgkin lymphoma allows omission of consolidation radiotherapy without a clinically relevant loss of efficacy. PET4-guided therapy could thereby reduce the proportion of patients at risk of the late effects of radiotherapy. FUNDING Deutsche Krebshilfe.
Collapse
Affiliation(s)
- Peter Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf, Faculty of Medicine and University of Cologne, Cologne, Germany; German Hodgkin Study Group, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Annette Plütschow
- Department I of Internal Medicine, Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf, Faculty of Medicine and University of Cologne, Cologne, Germany; German Hodgkin Study Group, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Richard Greil
- Illrd Medical Department, Paracelsus Medical University, Salzburg Austria; Salzburg Cancer Research Institute and AGMT, Salzburg, Austria
| | - Julia Meissner
- Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Max S Topp
- Medizinische Klinik Und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | | | - Judith Dierlamm
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Julia Thiemer
- Department of Hematology and Oncology, Klinikum der Philipps-Universität Marburg, Marburg, Germany
| | - Martin Sökler
- Innere Medizin II, University Hospital Tübingen, Tübingen, Germany
| | - Andrea Kerkhoff
- Medizinische Klinik A, University Hospital Münster, Münster, Germany
| | - Miriam Ahlborn
- Medizinische Klinik III, Städtisches Klinikum Braunschweig, Braunschweig, Germany
| | - Teresa V Halbsguth
- Department of Medicine II, Hematology/Oncology, Goethe University Hospital, Frankfurt, Frankfurt, Germany
| | - Sonja Martin
- Department of Hematology and Oncology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Ulrich Keller
- Klinikum rechts der Isar der TU München, München, Germany
| | - Stefan Balabanov
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Institute of Medical Oncology, Bern, Switzerland
| | - Martin Vogelhuber
- Klinik und Poliklinik für Innere Medizin III, Universitätsklinik Regensburg, Regensburg, Germany
| | - Andreas Hüttmann
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Martin Wilhelm
- Klinikum Nurnberg and Paracelsus Medical University, Nurnberg, Germany
| | - Josée M Zijlstra
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Alden Moccia
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Georg Kuhnert
- Department of Nuclear Medicine, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Paul J Bröckelmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf, Faculty of Medicine and University of Cologne, Cologne, Germany; German Hodgkin Study Group, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Bastian von Tresckow
- Department I of Internal Medicine, Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf, Faculty of Medicine and University of Cologne, Cologne, Germany; German Hodgkin Study Group, Faculty of Medicine and University of Cologne, Cologne, Germany; Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Fuchs
- Department I of Internal Medicine, Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf, Faculty of Medicine and University of Cologne, Cologne, Germany; German Hodgkin Study Group, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Beate Klimm
- Klinik für Hämatologie, Onkologie und Gastroenterologie, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius Maximilian University of Würzburg and Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Hans Eich
- Department of Radiotherapy, University Hospital Münster, Münster, Germany
| | - Christian Baues
- Department of Radiooncology and Cyberknife Center, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Simone Marnitz
- Department of Radiooncology and Cyberknife Center, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Volker Diehl
- German Hodgkin Study Group, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, Faculty of Medicine and University of Cologne, Cologne, Germany
| | - Andreas Engert
- Department I of Internal Medicine, Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf, Faculty of Medicine and University of Cologne, Cologne, Germany; German Hodgkin Study Group, Faculty of Medicine and University of Cologne, Cologne, Germany.
| |
Collapse
|
148
|
|
149
|
Haanen JB, Blank CU. Prognostic and predictive role of the tumor immune landscape. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2021; 64:143-151. [PMID: 32421286 DOI: 10.23736/s1824-4785.20.03255-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Cancer immunotherapy utilizing immune checkpoint inhibitors, has become mainstay for a growing number of cancer types. However, in general, the objective response rate and survival is only improved in a minority of cancer patients. Many of the biomarkers to select patients that either benefit or do not, have next to being predictive also prognostic value, are continuous instead of categorical, and are therefore difficult to use in daily practice. Currently, PD-L1 by immunohistochemical staining is the only approved biomarker to select metastatic non-small cell lung cancer patients for single agent pembrolizumab treatment. This may be extended with PD-L1 IHC staining in other cancer types, with different assays and different cut-offs. Other biomarkers, such as tumor mutational burden (TMB) by either whole exome sequencing or by targeted panel sequencing, gene expression profiling (GEP) either by NanoString or another RNA based sequencing test are under development. Assessment of immune infiltrates by multiplex immunohistochemistry or immunofluorescence also requires further validation. In the end, some of these tests may need to be combined in order to have the highest positive and negative predictive value and qualify as a reliable test for patient selection. The complexity and current status of biomarker research in immuno-oncology is outlined in this review.
Collapse
Affiliation(s)
- John B Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands -
| | - Christian U Blank
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
150
|
Topical and Systemic Formulation Options for Cutaneous T Cell Lymphomas. Pharmaceutics 2021; 13:pharmaceutics13020200. [PMID: 33540765 PMCID: PMC7913115 DOI: 10.3390/pharmaceutics13020200] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 11/17/2022] Open
Abstract
Although various anti-cutaneous T-cell lymphoma (CTCL) therapies are available for clinical use, appropriate chemotherapy lines for the treatment of CTCLs have yet to be established. Therefore, to date, various clinical trials for the treatment of advanced CTCLs are ongoing. In this review, we evaluate the therapeutic options that are available in clinical practice for treatment of early- and advanced-stage CTCLs (targeted therapies, histone deacetylase (HDAC) inhibitors, retinoids, interferons, cytotoxic drugs, etc.). We also examine clinical trials of novel regimens for the treatment of CTCLs.
Collapse
|