1
|
Takahashi H, Ito S, Nakanishi Y, Miura K, Nishimaki H, Nakagawa M, Otake S, Hamada T, Koike T, Iizuka K, Masuda S, Nakayama T, Shimizu T, Ishibashi N, Kogure H, Nakamura H. Topological importance of CD8 + T-cell enrichment in the tumor microenvironment of classic Hodgkin lymphoma. Ann Hematol 2025:10.1007/s00277-025-06189-1. [PMID: 39820429 DOI: 10.1007/s00277-025-06189-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Classic Hodgkin lymphoma (CHL) histologically consists of Hodgkin Reed-Sternberg (HRS) cells and the tumor microenvironment (TME), but the relationship between TME characteristics and clinical features of CHL remains unclear. We aimed to investigate the effects of the TME structure on the outcomes of patients with CHL. We performed a high-throughput analysis of HRS cells and their topological relationship with the reactive immune cells in the TME. After multiplexed immunofluorescence labeling against CD4, CD8, CD30, CD68, CD163, PD-1, and PD-L1, visual images were analyzed. Phenotypes were assigned to all reactive cells, such as CD4+ and CD8+ T-cells and macrophages. Since the densities of PD1+/CD4+ T-cells, CD8+ T-cells, and PD-L1+ macrophages were significantly higher in the area < 60 μm than in the area < 120 μm from each HRS cell in 45 tissue samples from 34 patients with CHL, we further analyzed the TME-component cells by focusing on the 60 μm radius in the initial samples. TMEs containing > 15 CD8+ T-cells were associated with a significantly better 3-year progression-free survival than those with ≤ 15 CD8+ T-cells (100% vs. 53%, p = 0.006). In comparison with TMEs containing ≤ 15 CD8+ T-cells, TMEs containing > 15 CD8+ T-cells had significantly more PD-L1- macrophages (mean 3 vs. 1 cell, p = 0.015) and fewer PD-1+/CD4+ T-cells (mean 16 vs. 28 cells, p = 0.036). Epstein-Barr virus positivity in HRS cells was significantly associated with a higher number of macrophages in the 60 μm radius area. In conclusion, the TME structure in patients with CHL can differ, enabling precision therapies.
Collapse
Affiliation(s)
- Hiromichi Takahashi
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Shun Ito
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Yoko Nakanishi
- Department of Pathology and Microbiology Division of Oncologic Pathology, Nihon University School of Medicine, Tokyo, Japan
| | - Katsuhiro Miura
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan.
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi City, Tokyo, 173-8610, Japan.
| | - Haruna Nishimaki
- Department of Pathology and Microbiology Division of Oncologic Pathology, Nihon University School of Medicine, Tokyo, Japan
| | - Masaru Nakagawa
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
- Present Address: Department of Hematology, Kasukabe Medical Center, Saitama, Japan
| | - Shimon Otake
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Takashi Hamada
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Takashi Koike
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Kazuhide Iizuka
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
- Department of Pathology and Microbiology Division of Laboratory Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Shinobu Masuda
- Department of Pathology and Microbiology Division of Oncologic Pathology, Nihon University School of Medicine, Tokyo, Japan
| | - Tomohiro Nakayama
- Department of Pathology and Microbiology Division of Laboratory Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tetsuo Shimizu
- Department of Medicine, Division of Respiratory Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Naoya Ishibashi
- Department of Radiology, Nihon University School of Medicine, Tokyo, Japan
| | - Hirofumi Kogure
- Department of Medicine Division of Gastroenterology and Hepatology, Nihon University School of Medicine, Tokyo, Japan
| | - Hideki Nakamura
- Department of Medicine Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Adashek JJ, Munoz JL, Kurzrock R. If it is a solid tumor target, then it may be a hematologic cancer target: Bridging the great divide. MED 2025; 6:100550. [PMID: 39689708 PMCID: PMC11725447 DOI: 10.1016/j.medj.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 12/19/2024]
Abstract
Tumor-agnostic US Food and Drug Administration approvals are transforming oncology. They include larotrectinib/entrectinib/repotrectinib (NTRK fusions), selpercatinib (RET fusions), dabrafenib/trametinib (BRAFV600E mutations), pembrolizumab/dostarlimab (microsatellite instability), pembrolizumab (high tumor mutational burden), and trastuzumab deruxtecan (HER2 3+ expression) (all solid cancers). Pemigatinib is approved for FGFR1-rearranged myeloid/lymphoid neoplasms. The genomically driven tissue-agnostic approach has a strong biological rationale (cancer is a disease of the genome), yields remarkably high response rates, and provides drug access to patients with an unmet need (rare/ultra-rare malignancies). Despite the solid tumor focus, both solid and hematologic cancers can harbor identical driver molecular abnormalities and respond to cognate therapies. For example, BRAFV600E and IDH1/2 mutations; ALK, FGFR, and NTRK fusions; PD-L1 amplification; and CD70 antigens are druggable in both solid and blood malignancies by gene-/immune-targeted therapies/chimeric antigen receptor T cells. Future biomarker-based tissue-agnostic basket studies/approvals should bridge the great divide and include both solid and hematologic cancers.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Javier L Munoz
- Department of Hematology, Mayo Clinic Arizona, Phoenix, AZ, USA.
| | - Razelle Kurzrock
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; WIN Consortium, Paris, France; University of Nebraska, Omaha, NE, USA.
| |
Collapse
|
3
|
Mella C, Tsarouhas P, Brockwell M, Ball HC. The Role of Chronic Inflammation in Pediatric Cancer. Cancers (Basel) 2025; 17:154. [PMID: 39796780 PMCID: PMC11719864 DOI: 10.3390/cancers17010154] [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: 12/04/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Inflammation plays a crucial role in wound healing and the host immune response following pathogenic invasion. However, unresolved chronic inflammation can result in tissue fibrosis and genetic alterations that contribute to the pathogenesis of human diseases such as cancer. Recent scientific advancements exploring the underlying mechanisms of malignant cellular transformations and cancer progression have exposed significant disparities between pediatric and adult-onset cancers. For instance, pediatric cancers tend to have lower mutational burdens and arise in actively developing tissues, where cell-cycle dysregulation leads to gene, chromosomal, and fusion gene development not seen in adult-onset counterparts. As such, scientific findings in adult cancers cannot be directly applied to pediatric cancers, where unique mutations and inherent etiologies remain poorly understood. Here, we review the role of chronic inflammation in processes of genetic and chromosomal instability, the tumor microenvironment, and immune response that result in pediatric tumorigenesis transformation and explore current and developing therapeutic interventions to maintain and/or restore inflammatory homeostasis.
Collapse
Affiliation(s)
- Christine Mella
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
| | - Panogiotis Tsarouhas
- Department of Biology, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA;
| | - Maximillian Brockwell
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
| | - Hope C. Ball
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
| |
Collapse
|
4
|
Lee J, Han MH, Baek DW. Successful Treatment of a Patient Presenting with Simultaneous Diffuse Large B-Cell Lymphoma and Hodgkin Lymphoma: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2025; 26:e945435. [PMID: 39748486 PMCID: PMC11706436 DOI: 10.12659/ajcr.945435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/22/2024] [Accepted: 11/13/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Simultaneously occuring diffuse large B-cell lymphoma (DLBCL) and Hodgkin lymphoma (HL) is extremely rare. Generally, patients with CD20-positive DLBCL receive rituximab, cyclophosphamide, vincristine, doxorubicin, prednisone (R-CHOP) regimen, while those with HL receive brentuximab vedotin, doxorubicin, vinblastine, dacarbazine (A-AVD) regimen as first-line therapy. Establishing a strategy for treating both lymphoma subtypes concurrently is thus very difficult. We report successful treatment of a patient simultaneously diagnosed with advanced DLBCL and HL. CASE REPORT A 20-year-old man visited the Hematology Department of Kyungpook National University Hospital after the diagnosis of germinal center B-cell DLBCL in the kidney and HL (nodular sclerosis type) in the neck lymph node. His DLBCL was classified as Ann Arbor stage IV with an International Prognostic Index score of 4, a high-risk group. Six cycles of R-CHOP therapy were planned, and central nervous system prophylaxis with intrathecalmethotrexate was added because of the high-risk features of central nervous system involvement. After completing 6 cycles of chemotherapy, without significant adverse events (Deauville score of 1), complete remission was confirmed. Then, the patient decided to undergo consolidative autologous stem cell transplantation (auto-SCT). He received busulfan, cyclophosphamide, and etoposide conditioning regimen, after which auto-SCT was conducted in April 2021. After auto-SCT, the patient was undergoing regular check-ups and doing well, without obvious disease relapse or specific symptoms. He maintained a disease-free status for 40 months to date. CONCLUSIONS Our case showed that R-CHOP regimen was effective not only for DLBCL but also for HL. Notably, consolidative upfront auto-SCT should be considered for a deeper response.
Collapse
Affiliation(s)
- Jungmin Lee
- Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Man Hoon Han
- Department of Pathology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Dong Won Baek
- Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
5
|
Lynch RC, Binkley MS. Rising to the Top: How Immune-Checkpoint Inhibitors are Changing the Landscape of Treatment for Classic Hodgkin Lymphoma. Semin Radiat Oncol 2025; 35:40-46. [PMID: 39672641 DOI: 10.1016/j.semradonc.2024.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024]
Abstract
Immune checkpoint inhibitors have transformed the future management of classic Hodgkin lymphoma (CHL) in the frontline and salvage settings. Both nivolumab and pembrolizumab have high efficacy in CHL allowing for reduction of chemotherapy and possibly the use of radiotherapy based on short-term follow-up. In this review article, we highlight recent trials enrolling patients with relapse and refractory classic Hodgkin lymphoma receiving monotherapy with PD-1 inhibitors as well as PD-1 and chemotherapy regimens. We also highlight frontline trials utilizing PD-1 inhibitors with chemotherapy with or without radiotherapy. Collectively, the role for radiotherapy in the frontline or salvage setting will become more defined with long-term follow-up of completed studies and results generated from future studies.
Collapse
Affiliation(s)
- Ryan C Lynch
- Division of Hematology/Oncology, University of Washington, Seattle, WA.
| | | |
Collapse
|
6
|
Paczkowska J, Tang M, Wright KT, Song L, Luu K, Shanmugam V, Welsh EL, Weirather JL, Besson N, Olszewski H, Porter BA, Pfaff KL, Redd RA, Cader FZ, Mandato E, Ouyang J, Calabretta E, Bai G, Lawton LN, Armand P, Rodig SJ, Liu XS, Shipp MA. Cancer-specific innate and adaptive immune rewiring drives resistance to PD-1 blockade in classic Hodgkin lymphoma. Nat Commun 2024; 15:10740. [PMID: 39737927 DOI: 10.1038/s41467-024-54512-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Hodgkin Reed-Sternberg (HRS) cells of classic Hodgkin lymphoma (cHL), like many solid tumors, elicit ineffective immune responses. However, patients with cHL are highly responsive to PD-1 blockade, which largely depends on HRS cell-specific retention of MHC class II and implicates CD4+ T cells and additional MHC class I-independent immune effectors. Here, we utilize single-cell RNA sequencing and spatial analysis to define shared circulating and microenvironmental features of the immune response to PD-1 blockade in cHL. Compared with non-responders, responding patients have more circulating CD4+ naïve and central memory T cells and B cells, as well as more diverse CD4+ T cell and B cell receptor repertoires. Importantly, a population of circulating and tumor-infiltrating IL1β+ monocytes/macrophages is detectable in patients with cHL but not healthy donors, and a proinflammatory, tumor-promoting signature of these circulating IL1β+ monocytes is associated with resistance to PD-1 blockade in cHL. Altogether, our findings reveal extensive immune rewiring and complementary roles of CD4+ T cells, B cells and IL1β+ monocytes in the response to PD-1 blockade and suggest that these features can be captured with a peripheral blood test.
Collapse
Affiliation(s)
- Julia Paczkowska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ming Tang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Astra Zeneca, Waltham, MA, USA
| | - Kyle T Wright
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Li Song
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Data Science, Dartmouth College, Hanover, NH, USA
| | - Kelsey Luu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- PathAI, Boston, MA, USA
| | - Vignesh Shanmugam
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emma L Welsh
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jason L Weirather
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Naomi Besson
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Harrison Olszewski
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Billie A Porter
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L Pfaff
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Robert A Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fathima Zumla Cader
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- AstraZeneca, City House, Cambridge, UK
| | - Elisa Mandato
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jing Ouyang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Mechanisms of Cancer Resistance Thematic Center, Bristol Myers Squibb, Cambridge, MA, USA
| | - Eleonora Calabretta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gali Bai
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Lee N Lawton
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xiaole Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- GV20 Therapeutics, LLC, Cambridge, MA, USA
| | - Margaret A Shipp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
7
|
Nakayama T, Takahama T, Chiba Y, Shiraishi N, Kawakami H, Yonesaka K, Nakagawa K, Hayashi H. Evaluation of immune checkpoint inhibitor efficacy for solid tumors with CD274 (PD-L1 gene) amplification identified by comprehensive genomic profiling: retrospective study based on a nationwide database. J Immunother Cancer 2024; 12:e010130. [PMID: 39694703 PMCID: PMC11667388 DOI: 10.1136/jitc-2024-010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Amplification of the programmed cell death-ligand 1 gene (CD274) is highly prevalent and associated with a high response rate to immune checkpoint inhibitors (ICIs) in lymphomas, and is also a potential biomarker for ICI treatment of solid tumors. However, the efficacy of ICIs for solid tumors with CD274 amplification identified by comprehensive genomic profiling (CGP) has been unclear. We here examined ICI efficacy for solid tumors with CD274 amplification identified by CGP in a national database. METHODS We retrospectively analyzed data from the Center for Cancer Genomics and Advanced Therapeutics database containing 60,155 CGP test results for individuals with solid tumors. Only clinical data from patients treated with ICIs alone (not those undergoing concomitant therapy with molecularly targeted or cytotoxic chemotherapeutic agents) were evaluated. We matched 48 patients in the CD274 amplification-positive group with 170 patients in the CD274 amplification-negative group in a 1:4 ratio based on tumor type, histology, treatment, and age. Overall survival (OS), time to next treatment (TTNT), and response rate were evaluated as treatment outcomes in the two groups. RESULTS OS was similar in the CD274-amplified and matched CD274-non-amplified groups (median of 22.1 vs 26.3 months, respectively; HR of 0.92 with a 95% CI of 0.55 to 1.54; p=0.075). TTNT tended to be longer in the CD274-amplified group than in the matched CD274-non-amplified group (median of 16.5 vs 14.0 months; HR of 0.63 with a 95% CI of 0.37 to 1.08; p=0.091). The objective response rate was 33.3% and 18.4% (difference of 14.9%, with a 95% CI of -0.2% to 31.6%), and the disease control rate was 63.9% and 41.1% (difference of 22.8%, with a 95% CI of 5.1% to 40.4%), in the CD274-amplified and matched CD274-non-amplified groups, respectively. CONCLUSIONS The number of patients with solid tumors positive for CD274 amplification in this analysis is the largest to date, and our results suggest that such gene amplification may be associated with the outcome of ICI treatment in such individuals. CD274 amplification identified by CGP may therefore be a predictor of ICI efficacy for solid tumors. TRIAL REGISTRATION NUMBER UMIN000029779.
Collapse
Affiliation(s)
- Tomohiro Nakayama
- Medical Oncology, Kishiwada City Hospital, Kishiwada, Japan
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Takayuki Takahama
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Genome Medical Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Naoki Shiraishi
- Genome Medical Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kimio Yonesaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Genome Medical Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Cancer Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
8
|
Santisteban Espejo A, Bernal-Florindo I, Montero-Pavon P, Perez-Requena J, Atienza-Cuevas L, Villalba-Fernandez A, Garcia-Rojo M. Whole slide imaging of tumour microenvironment in classical Hodgkin's lymphoma: development of a clinical prediction model based on programmed death-ligand 1 and tumorous Reed-Sternberg cells. J Clin Pathol 2024; 78:11-18. [PMID: 37977655 DOI: 10.1136/jcp-2023-209097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/29/2023] [Indexed: 11/19/2023]
Abstract
AIMS The prognostic impact of programmed death-ligand 1 (PD-L1) cells in classic Hodgkin lymphoma (cHL) tumour microenvironment remains undefined. METHODS Model development via Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis guidelines were followed. PD-L1+ and CD30+ tumoral Reed-Sternberg cells were quantified through whole slide imaging and digital image analysis in 155 digital histopathological slides of cHL. Univariate and multivariate survival analyses were performed. The analyses were reproduced for patients with advanced stages (IIB, III and IV) using the Advanced-stage cHL International Prognostic Index. RESULTS The PD-L1/CD30 ratio was statistically significantly associated with survival outcomes. Patients with a PD-L1/CD30 ratio above 47.1 presented a shorter overall survival (mean OS: 53.7 months; 95% CI: 28.7 to 78.7) in comparison with patients below this threshold (mean OS: 105.4 months; 95% CI: 89.6 to 121.3) (p=0.04). When adjusted for covariates, the PD-L1/CD30 ratio retained prognostic impact, both for the OS (HR: 1.005; 95% CI: 1.002 to 1.008; p=0.000) and the progression-free survival (HR: 3.442; 95% CI: 1.045 to 11.340; p=0.04) in a clinical and histopathological multivariate model including the male sex (HR: 3.551; 95% CI: 0.986 to 12.786; p=0.05), a percentage of tumoral cells ≥10.1% (HR: 1.044; 95% CI: 1.003 to 1.087; p=0.03) and high risk International Prognostic Score (≥3 points) (HR: 6.453; 95% CI: 1.970 to 21.134; p=0.002). CONCLUSIONS The PD-L1/CD30 ratio identifies a group of cHL patients with an increased risk of treatment failure. Its clinical application can be performed as it constitutes an easy to implement pathological information in the diagnostic work-up of patients with cHL.
Collapse
Affiliation(s)
- Antonio Santisteban Espejo
- Department of Pathology, Puerta del Mar University Hospital, Cadiz, Spain
- Deparment of Medicine, Faculty of Medicine, University of Cadiz, Cadiz, Spain
- Institute of Research and Biomedical Innovation of Cadiz, INiBICA, Cadiz, Spain
| | - Irene Bernal-Florindo
- Institute of Research and Biomedical Innovation of Cadiz, INiBICA, Cadiz, Spain
- Department of Pathology, Jerez de la Frontera University Hospital, Jerez de la Frontera, Spain
| | - Pedro Montero-Pavon
- Department of Pathology, Jerez de la Frontera University Hospital, Jerez de la Frontera, Spain
| | - Jose Perez-Requena
- Department of Pathology, Puerta del Mar University Hospital, Cadiz, Spain
| | - Lidia Atienza-Cuevas
- Department of Pathology, Puerta del Mar University Hospital, Cadiz, Spain
- Institute of Research and Biomedical Innovation of Cadiz, INiBICA, Cadiz, Spain
| | | | - Marcial Garcia-Rojo
- Institute of Research and Biomedical Innovation of Cadiz, INiBICA, Cadiz, Spain
- Department of Pathology, Jerez de la Frontera University Hospital, Jerez de la Frontera, Spain
| |
Collapse
|
9
|
Ullrich F, Bröckelmann PJ, Turki AT, Khan AM, Chiru ED, Vetter M, von Tresckow B, Wirth R, Cordoba R, Ortiz-Maldonado V, Fülöp T, Neuendorff NR. Impact of immunological aging on T cell-mediated therapies in older adults with multiple myeloma and lymphoma. J Immunother Cancer 2024; 12:e009462. [PMID: 39622581 PMCID: PMC11624774 DOI: 10.1136/jitc-2024-009462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/24/2024] [Indexed: 12/09/2024] Open
Abstract
The treatment landscape for lymphoma and multiple myeloma, which disproportionally affect older adults, has been transformed by the advent of T cell-mediated immunotherapies, including immune checkpoint inhibition, T cell-engaging bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapy, during the last decade. These treatment modalities re-enable the patient's own immune system to combat malignant cells and offer the potential for sustained remissions and cure for various diseases.Age profoundly affects the physiological function of the immune system. The process of biological aging is largely driven by inflammatory signaling, which is reciprocally fueled by aging-related alterations of physiology and metabolism. In the T cell compartment, aging contributes to T cell senescence and exhaustion, increased abundance of terminally differentiated cells, a corresponding attrition in naïve T cell numbers, and a decrease in the breadth of the receptor repertoire. Furthermore, inflammatory signaling drives aging-related pathologies and contributes to frailty in older individuals. Thus, there is growing evidence of biological aging modulating the efficacy and toxicity of T cell-mediated immunotherapies.Here, we review the available evidence from biological and clinical studies focusing on the relationship between T cell-mediated treatment of hematologic malignancies and age. We discuss biological features potentially impacting clinical outcomes in various scenarios, and potential strategies to improve the safety and efficacy of immune checkpoint inhibitors, T cell-engaging bispecific antibodies, and CAR-T cell therapy in older patients.
Collapse
Affiliation(s)
- Fabian Ullrich
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Paul J Bröckelmann
- Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD) and German Hodgkin Study Group (GHSG), University of Cologne, Cologne, Germany
- Max Planck Institute for Biology of Ageing, Cologne, Nordrhein-Westfalen, Germany
| | - Amin T Turki
- Department of Hematology and Oncology, University Hospital Marien Hospital Herne, Herne, Nordrhein-Westfalen, Germany
- Institute for Artificial Intelligence in Medicine, University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Abdullah M Khan
- Division of Hematology, The Ohio State University Comprehensive Cancer Center Arthur G James Cancer Hospital and Richard J Solove Research Institute, Columbus, Ohio, USA
| | - Elena-Diana Chiru
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Marcus Vetter
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Rainer Wirth
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| | - Raul Cordoba
- Department of Hematology, Lymphoma Unit, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | - Valentín Ortiz-Maldonado
- Department of Hematology, Oncoimmunotherapy Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Tamas Fülöp
- Department of Medicine, Division of Geriatrics, Research Center on Aging, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Nina Rosa Neuendorff
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| |
Collapse
|
10
|
Ansell SM. Hodgkin lymphoma: 2025 update on diagnosis, risk-stratification, and management. Am J Hematol 2024; 99:2367-2378. [PMID: 39239794 DOI: 10.1002/ajh.27470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
DISEASE OVERVIEW Hodgkin lymphoma (HL) is an uncommon B-cell lymphoid malignancy affecting 8570 new patients annually and representing ~10% of all lymphomas in the United States. DIAGNOSIS HL is composed of two distinct disease entities: classical HL and nodular lymphocyte predominant HL (also called nodular lymphocyte predominant B-cell lymphoma). Nodular sclerosis, mixed cellularity, lymphocyte depletion, and lymphocyte-rich HL are subgroups of classical HL. RISK STRATIFICATION An accurate assessment of the stage of disease in patients with HL is critical for the selection of the appropriate therapy. Prognostic models that identify patients at low or high risk for recurrence, as well as the response to therapy as determined by positron emission tomography (PET) scan, are used to optimize therapy. RISK-ADAPTED THERAPY Initial therapy for HL patients is based on the histology of the disease, the anatomical stage and the presence of poor prognostic features. Patients with early-stage disease are typically treated with combined modality strategies utilizing abbreviated courses of combination chemotherapy followed by involved-field radiation therapy, whereas those with advanced stage disease receive a longer course of chemotherapy often without radiation therapy. However, newer agents including brentuximab vedotin and anti-PD-1 antibodies are now standardly incorporated into frontline therapy. MANAGEMENT OF RELAPSED/REFRACTORY DISEASE High-dose chemotherapy (HDCT) followed by an autologous stem cell transplant (ASCT) is the standard of care for most patients who relapse following initial therapy. For patients who fail HDCT with ASCT, brentuximab vedotin, PD-1 blockade, non-myeloablative allogeneic transplant or participation in a clinical trial should be considered.
Collapse
Affiliation(s)
- Stephen M Ansell
- Dorotha W. and Grant L. Sundquist Professor in Hematologic Malignancies Research Chair, Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Bisig B, Lefort K, Carras S, de Leval L. Clinical use of circulating tumor DNA analysis in patients with lymphoma. Hum Pathol 2024:105679. [PMID: 39491629 DOI: 10.1016/j.humpath.2024.105679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The analysis of circulating tumor DNA (ctDNA) in liquid biopsy specimens has an established role for the detection of predictive molecular alterations and acquired resistance mutations in several tumors. The low-invasiveness of this approach allows for repeated sampling and dynamic monitoring of disease evolution. Originating from the entire body tumor bulk, plasma-derived ctDNA reflects intra- and interlesional genetic heterogeneity. In the management of lymphoma patients, ctDNA quantification at various timepoints of the patient's clinical history is emerging as a complementary tool that may improve risk stratification, assessment of treatment response and early relapse detection during follow-up, most prominently in patients with diffuse large B-cell lymphoma or classic Hodgkin lymphoma. While liquid biopsies have not yet entered standard-of-care treatment protocols in these settings, several trials have provided evidence that at least a subset of lymphoma patients may benefit from the introduction of liquid biopsies into daily clinical care. In parallel, continuous technological developments have enabled highly sensitive ctDNA assessment methods, which span from locus-specific techniques identifying single hotspot mutations, to sequencing panels and genome-wide approaches that explore broader genetic and epigenetic alterations. Here, we provide an overview of current methods and ongoing technical developments for ctDNA evaluation. We also summarize the most important data from a selection of clinical studies that have explored the clinical use of ctDNA in several lymphoma entities.
Collapse
Affiliation(s)
- Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Karine Lefort
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Sylvain Carras
- Institute for Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Department of Molecular Biology and Department of Oncohematology, University Hospital Grenoble and University Grenoble Alpes, Grenoble, France
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| |
Collapse
|
12
|
Gunawardana J, Law SC, Sabdia MB, Fennell É, Hennessy A, Leahy CI, Murray PG, Bednarska K, Brosda S, Trotman J, Berkahn L, Zaharia A, Birch S, Burgess M, Talaulikar D, Lee JN, Jude E, Hawkes EA, Jain S, Nath K, Snell C, Swain F, Tobin JWD, Keane C, Shanavas M, Blyth E, Steidl C, Savage K, Farinha P, Boyle M, Meissner B, Green MR, Vega F, Gandhi MK. Intra-tumoral and peripheral blood TIGIT and PD-1 as immune biomarkers in nodular lymphocyte predominant Hodgkin lymphoma. Am J Hematol 2024; 99:2096-2107. [PMID: 39152767 PMCID: PMC11469944 DOI: 10.1002/ajh.27459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/28/2024] [Indexed: 08/19/2024]
Abstract
In classical Hodgkin lymphoma (cHL), responsiveness to immune-checkpoint blockade (ICB) is associated with specific tumor microenvironment (TME) and peripheral blood features. The role of ICB in nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is not established. To gain insights into its potential in NLPHL, we compared TME and peripheral blood signatures between HLs using an integrative multiomic analysis. A discovery/validation approach in 121 NLPHL and 114 cHL patients highlighted >2-fold enrichment in programmed cell death-1 (PD-1) and T-cell Ig and ITIM domain (TIGIT) gene expression for NLPHL versus cHL. Multiplex imaging showed marked increase in intra-tumoral protein expression of PD-1+ (and/or TIGIT+) CD4+ T-cells and PD-1+CD8+ T-cells in NLPHL compared to cHL. This included T-cells that rosetted with lymphocyte predominant (LP) and Hodgkin Reed-Sternberg (HRS) cells. In NLPHL, intra-tumoral PD-1+CD4+ T-cells frequently expressed TCF-1, a marker of heightened T-cell response to ICB. The peripheral blood signatures between HLs were also distinct, with higher levels of PD-1+TIGIT+ in TH1, TH2, and regulatory CD4+ T-cells in NLPHL versus cHL. Circulating PD-1+CD4+ had high levels of TCF-1. Notably, in both lymphomas, highly expanded populations of clonal TIGIT+PD-1+CD4+ and TIGIT+PD-1+CD8+ T-cells in the blood were also present in the TME, indicating that immune-checkpoint expressing T-cells circulated between intra-tumoral and blood compartments. In in vitro assays, ICB was capable of reducing rosette formation around LP and HRS cells, suggesting that disruption of rosetting may be a mechanism of action of ICB in HL. Overall, results indicate that further evaluation of ICB is warranted in NLPHL.
Collapse
Affiliation(s)
- Jay Gunawardana
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Soi C. Law
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Muhammed B. Sabdia
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Éanna Fennell
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
| | - Aoife Hennessy
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
| | - Ciara I. Leahy
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
| | - Paul G. Murray
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
- Royal College of Surgeons Ireland, Adliya, Bahrain
| | - Karolina Bednarska
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Sandra Brosda
- Frazer Institute, University of Queensland, Translational Research Institute Brisbane, Australia
| | - Judith Trotman
- Concord Repatriation General Hospital, University of Sydney, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW
| | - Leanne Berkahn
- Department of Haematology, Auckland City Hospital, Auckland, New Zealand
| | - Andreea Zaharia
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Simone Birch
- Princess Alexandra Hospital, Brisbane, Australia
| | - Melinda Burgess
- School of Medicine, Limerick Digital Cancer Research Centre, Health Research Institute and Bernal Institute, University of Limerick, Limerick, Ireland
- Princess Alexandra Hospital, Brisbane, Australia
| | - Dipti Talaulikar
- Haematology Translational Research Unit, ACT Pathology, Canberra Health Services, Canberra, Australia
- College of Health and Medicine, Australian National University, Canberra, Australia
| | - Justina N. Lee
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | | | - Eliza A. Hawkes
- Olivia Newton John Cancer Research and Wellness Centre, Austin Health, Melbourne, Australia
- Transfusion Research Unit, School of Public Health and Preventative Medicine, Monash University, Melbourne Australia
| | - Sanjiv Jain
- Anatomical Pathology Department, The Canberra Hospital, Canberra, Australia
| | - Karthik Nath
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
- Memorial Sloan Kettering Cancer Center, NY, USA
| | - Cameron Snell
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Mater Pathology, Brisbane, Australia
| | - Fiona Swain
- Royal College of Surgeons Ireland, Adliya, Bahrain
- Princess Alexandra Hospital, Brisbane, Australia
| | - Joshua W. D. Tobin
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
| | - Colm Keane
- Frazer Institute, University of Queensland, Translational Research Institute Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
| | - Mohamed Shanavas
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Emily Blyth
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW
- Department of Haematology, Westmead Hospital, Westmead, NSW, Australia
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW
| | | | - Kerry Savage
- British Columbia Cancer Agency, Vancouver, Canada
| | | | | | | | | | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maher K. Gandhi
- Blood Cancer Research Group, Mater Research, University of Queensland, Translational Research Institute, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
13
|
Nie J, Wang C, Zheng L, Liu Y, Wang C, Chang Y, Hu Y, Guo B, Pan Y, Yang Q, Hu X, Han W. Epigenetic agents plus anti-PD-1 reprogram the tumor microenvironment and restore antitumor efficacy in Hodgkin lymphoma. Blood 2024; 144:1936-1950. [PMID: 39093981 DOI: 10.1182/blood.2024024487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024] Open
Abstract
ABSTRACT DNA methyltransferase inhibitor decitabine plus anti-programmed cell death 1 (DP) therapy was effective in relapsed/refractory classic Hodgkin lymphoma (cHL). However, a subset of patients experienced primary resistance or relapse/progression after DP therapy. In this study, we evaluated the efficacy and safety of a triplet regimen consisting of the histone deacetylase inhibitor chidamide, decitabine, and anti-PD-1 camrelizumab (CDP) in 52 patients who previously received DP therapy. CDP treatment was well tolerated and resulted in an objective response rate of 94% (95% confidence interval [CI], 84-99), with 50% (95% CI, 36-64) of patients achieving complete response (CR). Notably, all patients who were recalcitrant to previous DP treatment exhibited therapeutic responses after CDP therapy, although their CR rate was lower than patients responsive to prior DP. Overall, the median progression-free survival was 29.4 months. Through single-cell RNA sequencing of pretreatment and on-treatment cHL tumor biopsy samples, we observed the heterogeneity of rare malignant Hodgkin Reed/Sternberg (HRS)-like cells. The classical CD30+ HRS-like cells interacted with abundant immunosuppressive IL21+CD4+ T helper cells, forming a positive feedback loop that supported their survival. While the CD30- HRS-like cell population showed potential resistance to anti-PD-1 immunotherapy. CDP treatment promoted the activation of diverse tumor-reactive CD8+ T cells and suppressed the proliferation of IL21+CD4+ T cells by inhibiting STAT1/3 signaling, thereby alleviating their immunosuppressive effects. These findings provide insights into the cHL microenvironment that contributes to anti-PD-1 resistance and highlight the therapeutic effectiveness of dual epi-immunotherapy in overcoming immunotherapy resistance. This trial was registered at www.clinicaltrials.gov as #NCT04233294.
Collapse
Affiliation(s)
- Jing Nie
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chunmeng Wang
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | | | - Yang Liu
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | | | - Yixin Chang
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yudi Hu
- Analytical Biosciences Limited, Beijing, China
| | - Bing Guo
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuting Pan
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Qingming Yang
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xueda Hu
- Analytical Biosciences Limited, Beijing, China
| | - Weidong Han
- Department of Biotherapeutics, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
- Changping Laboratory, Beijing, China
| |
Collapse
|
14
|
Li Y, Wu Y, Cao S, Yu B, Zhang Q, Xia Z, Cao J, Lv F, Chen GL. PD-1 inhibitors plus chemotherapy for refractory EBV-positive DLBCL: a retrospective analysis. Blood Res 2024; 59:36. [PMID: 39475995 PMCID: PMC11525352 DOI: 10.1007/s44313-024-00042-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Immunochemotherapy has demonstrated a promising efficacy for a variety of B-cell lymphoma but has limited efficacy for Epstein-Barr virus-positive (EBV +) diffuse large B-cell lymphoma (DLBCL) that is refractory or relapsed to conventional chemotherapy regimens. Considering higher programmed death-ligand 1 (PD-L1) expression in the subset of patients with DLBCL with positive EBV, we speculated that PD-1 inhibitors plus chemotherapy may be an alternative regimen in patients with refractory/relapsed EBV + DLBCL. METHODS This retrospective study included six adult patients diagnosed with refractory EBV + DLBCL resistant to first-line immunochemotherapy regimens (R-CHOP). These patients received PD-1 inhibitors plus chemotherapy as second-line treatment. RESULTS The final analysis included six patients (four men and two women (median age, 50 years; range, 39-83 years)). Four patients were diagnosed with Epstein-Barr virus (EBV) + DLBCL, and two had DLBCL associated with chronic inflammation. Over a median follow-up of 20 months (range, 2-31 months), the objective response rate was 83% (5/6) and the complete remission rate was 67% (4/6). No severe immune-related adverse reactions occurred, and only a mild rash was reported, which did not necessitate the discontinuation of therapy. CONCLUSION The combination of PD-1 inhibitors and chemotherapy offers promising results as a second-line treatment for patients with refractory EBV + DLBCL that is resistant to first-line immunochemotherapy regimens. These preliminary findings warrant further investigation in larger clinical trials to validate the efficacy and safety of this therapeutic approach.
Collapse
Affiliation(s)
- Youli Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center Xiamen Hospital, Xiamen, 361026, P.R. China
| | - Yonghe Wu
- Department of Pathology, Fujian Province, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, P.R. China
| | - Sufen Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
- Department of Nursing, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
| | - Baohua Yu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
| | - Qunling Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China
| | - Zuguang Xia
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China
| | - Junning Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China.
| | - Fangfang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China.
| | - Guang-Liang Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P.R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China.
| |
Collapse
|
15
|
Daunov M, van Besien K. High-Dose Chemotherapy and Autologous or Allogeneic Transplantation in Aggressive B-Cell Lymphoma-Is There Still a Role? Cells 2024; 13:1780. [PMID: 39513887 PMCID: PMC11545473 DOI: 10.3390/cells13211780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Novel therapies such as CAR-T, BTK inhibitors and PD-1 inhibitors have changed the management of aggressive B-cell lymphomas. Nonetheless, these novel therapies have their own risk of late toxicities including second malignancies. They also create a subgroup of patients with relapse, treatment failure, or indefinite maintenance. We discuss the current role of autologous and allogeneic stem cell transplantation in this context. In patients with recurrent diffuse large B-cell lymphoma, CAR-T cell treatment has largely replaced autologous transplant. Autologous transplant should be considered in patients with late relapses and in selected patients with T-cell-rich B-cell lymphoma, where CAR-T cell therapy may be less effective. It also remains the treatment of choice for consolidation of patients with primary CNS lymphoma. In mantle cell lymphoma, intensive chemotherapy combined with BTK inhibitors and rituximab results in excellent outcomes, and the role of autologous transplantation is declining. In Hodgkin's lymphoma, autologous transplant consolidation remains the standard of care for patients who failed initial chemotherapy. Allogeneic transplantation has lower relapse rates but more complications and higher non-relapse mortality than autologous transplantation. It is usually reserved for patients who fail autologous transplantation or in whom autologous stem cells cannot be collected. It may also have an important role in patients who fail CAR-T therapies. The increasing complexity of care and evolving sequencing of therapies for patients with aggressive B-cell lymphomas only emphasizes the importance of appropriate patient selection and optimal timing of stem cell transplantation.
Collapse
Affiliation(s)
| | - Koen van Besien
- University Hospitals, Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA;
| |
Collapse
|
16
|
Nayak RK, Aiello M, Maldonado LM, Clark TY, Buchwald ZS, Chang A. Impact of race, ethnicity, and social determinants on outcomes following immune checkpoint therapy. J Immunother Cancer 2024; 12:e010116. [PMID: 39461882 PMCID: PMC11529590 DOI: 10.1136/jitc-2024-010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/03/2024] [Indexed: 10/29/2024] Open
Abstract
Immune checkpoint blockade (ICB) therapies are one of the greatest advances in the history of cancer care and are now commonly used in the management of many different malignancies. However, much remains unknown about the factors that affect the efficacy and side effect profile of these agents. This review delves into the published literature that evaluates the intricate interplay between race, age, gender, and social determinants in shaping outcomes following ICB across solid tumors and hematologic malignancies. We examine the pivotal phase 2 and 3 trials to evaluate the demographics of participants and outcomes based on these variables, if reported. Most, but not all, trials reported some basic demographic information like age, sex, race, ethnicity, and/or geographic area for enrollment. Clinically relevant biological markers that could affect ICB outcomes such as obesity or markers of social determinants of health were largely not reported. Trials were generally representative for men and women based on expected prevalence for a given malignancy, but often under-represented non-white participants and rarely enrolled patients from the global south. Subgroup analyses were conducted in many ICB trials for solid malignancies, but rarely conducted for hematologic malignancies. These analyses largely showed similar qualitative benefit across subgroups, but adverse events were rarely reported by subgroup. This review adds to our understanding of the populations that these clinical trials have studied and highlight the urgent need to redouble our efforts at increasing the diversity of the population in future ICB trials.
Collapse
Affiliation(s)
- Rahul K Nayak
- Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Meili Aiello
- Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | | | - Tarralyn Y Clark
- Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Zachary S Buchwald
- Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Andres Chang
- Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Knecht H, Petrogiannis-Haliotis T, Louis S, Mai S. 3D-Q-FISH/Telomere/TRF2 Nanotechnology Identifies a Progressively Disturbed Telomere/Shelterin/Lamin AC Complex as the Common Pathogenic, Molecular/Spatial Denominator of Classical Hodgkin Lymphoma. Cells 2024; 13:1748. [PMID: 39513855 PMCID: PMC11545283 DOI: 10.3390/cells13211748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The bi- or multinucleated Reed-Sternberg cell (RS) is the diagnostic cornerstone of Epstein-Barr Virus (EBV)-positive and EBV-negative classical Hodgkin lymphoma (cHL). cHL is a germinal center (GC)-derived B-cell disease. Hodgkin cells (H) are the mononuclear precursors of RS. An experimental model has to fulfill three conditions to qualify as common pathogenic denominator: (i) to be of GC-derived B-cell origin, (ii) to be EBV-negative to avoid EBV latency III expression and (iii) to support permanent EBV-encoded oncogenic latent membrane protein (LMP1) expression upon induction. These conditions are unified in the EBV-, diffuse large B-Cell lymphoma (DLBCL) cell line BJAB-tTA-LMP1. 3D reconstructive nanotechnology revealed spatial, quantitative and qualitative disturbance of telomere/shelterin interactions in mononuclear H-like cells, with further progression during transition to RS-like cells, including progressive complexity of the karyotype with every mitotic cycle, due to BBF (breakage/bridge/fusion) events. The findings of this model were confirmed in diagnostic patient samples and correlate with clinical outcomes. Moreover, in vitro, significant disturbance of the lamin AC/telomere interaction progressively occurred. In summary, our research over the past three decades identified cHL as the first lymphoid malignancy driven by a disturbed telomere/shelterin/lamin AC interaction, generating the diagnostic RS. Our findings may act as trailblazer for tailored therapies in refractory cHL.
Collapse
Affiliation(s)
- Hans Knecht
- Division of Hematology, Department of Medicine, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | | | - Sherif Louis
- Telo Genomics Corp., Ontario, ON M5G 1L7, Canada;
| | - Sabine Mai
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada;
| |
Collapse
|
18
|
Cashen AF. The evolving role of checkpoint inhibitors in the treatment of Hodgkin lymphoma. Front Oncol 2024; 14:1392653. [PMID: 39502311 PMCID: PMC11534676 DOI: 10.3389/fonc.2024.1392653] [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: 02/27/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Since their initial approval as single agent therapy for multiply relapsed/refractory Hodgkin lymphoma (HL), the PD-1 inhibitors nivolumab and pembrolizumab have been incorporated into second-line salvage regimens, and they are being investigated in upfront therapy of newly diagnosed patients. As second-line therapy in combination with brentuximab vedotin or multi-agent chemotherapy, nivolumab and pembrolizumab provide high complete remission rates and durable progression-free survival after consolidative autologous stem cell transplant. Incorporation of these agents into frontline chemotherapy regimens is feasible, and early results from a Phase III trial of nivolumab-AVD compare favorably with the existing standard for advanced stage HL, brentuximab vedotin plus AVD. As nivolumab and pembrolizumab move into earlier lines of HL therapy, open research questions include the efficacy of checkpoint inhibitor regimens in patients who relapse after frontline exposure to nivolumab or pembrolizumab; the selection of patients with relapsed HL who can achieve durable remissions without autologous stem cell transplant; and the efficacy of the PD-1 inhibitors in the frontline therapy of patients with early stage Hodgkin lymphoma.
Collapse
Affiliation(s)
- Amanda F. Cashen
- Washington University School of Medicine, Siteman Cancer Center, St.
Louis, MO, United States
| |
Collapse
|
19
|
Herrera AF, LeBlanc M, Castellino SM, Li H, Rutherford SC, Evens AM, Davison K, Punnett A, Parsons SK, Ahmed S, Casulo C, Bartlett NL, Tuscano JM, Mei MG, Hess BT, Jacobs R, Saeed H, Torka P, Hu B, Moskowitz C, Kaur S, Goyal G, Forlenza C, Doan A, Lamble A, Kumar P, Chowdury S, Brinker B, Sharma N, Singh A, Blum KA, Perry AM, Kovach A, Hodgson D, Constine LS, Shields LK, Prica A, Dillon H, Little RF, Shipp MA, Crump M, Kahl B, Leonard JP, Smith SM, Song JY, Kelly KM, Friedberg JW. Nivolumab+AVD in Advanced-Stage Classic Hodgkin's Lymphoma. N Engl J Med 2024; 391:1379-1389. [PMID: 39413375 PMCID: PMC11488644 DOI: 10.1056/nejmoa2405888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
BACKGROUND Incorporating brentuximab vedotin into the treatment of advanced-stage classic Hodgkin's lymphoma improves outcomes in adult and pediatric patients. However, brentuximab vedotin increases the toxic effects of treatment in adults, more than half of pediatric patients who receive the drug undergo consolidative radiation, and relapse remains a challenge. Programmed death 1 blockade is effective in Hodgkin's lymphoma, including in preliminary studies involving previously untreated patients. METHODS We conducted a phase 3, multicenter, open-label, randomized trial involving patients at least 12 years of age with stage III or IV newly diagnosed Hodgkin's lymphoma. Patients were randomly assigned to receive brentuximab vedotin with doxorubicin, vinblastine, and dacarbazine (BV+AVD) or nivolumab with doxorubicin, vinblastine, and dacarbazine (N+AVD). Prespecified patients could receive radiation therapy directed to residual metabolically active lesions. The primary end point was progression-free survival, defined as the time from randomization to the first observation of progressive disease or death from any cause. RESULTS Of 994 patients who underwent randomization, 970 were included in the intention-to-treat population for efficacy analyses. At the second planned interim analysis, with a median follow-up of 12.1 months, the threshold for efficacy was crossed, indicating that N+AVD significantly improved progression-free survival as compared with BV+AVD (hazard ratio for disease progression or death, 0.48; 99% confidence interval [CI], 0.27 to 0.87; two-sided P = 0.001). Owing to the short follow-up time, we repeated the analysis with longer follow-up; with a median follow-up of 2.1 years (range, 0 to 4.2 years), the 2-year progression-free survival was 92% (95% CI, 89 to 94) with N+AVD, as compared with 83% (95% CI, 79 to 86) with BV+AVD (hazard ratio for disease progression or death, 0.45; 95% CI, 0.30 to 0.65). Overall, 7 patients received radiation therapy. Immune-related adverse events were infrequent with nivolumab; brentuximab vedotin was associated with more treatment discontinuation. CONCLUSIONS N+AVD resulted in longer progression-free survival than BV+AVD in adolescents and adults with stage III or IV advanced-stage classic Hodgkin's lymphoma and had a better side-effect profile. (Funded by the National Cancer Institute of the National Institutes of Health and others; S1826 ClinicalTrials.gov number, NCT03907488.).
Collapse
Affiliation(s)
| | | | - Sharon M. Castellino
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University Hospital/Winship Cancer Institute, Atlanta, GA
| | - Hongli Li
- SWOG Statistics and Data Management Center, Seattle, WA
| | | | | | - Kelly Davison
- McGill University Health Centre, Montreal, Quebec, CAN
| | | | - Susan K. Parsons
- Reid R. Sacco AYA Cancer Program, Tufts Medical Center, Boston, MA
| | | | - Carla Casulo
- Wilmot Cancer Institute, University of Rochester, Rochester, NY
| | | | | | | | - Brian T. Hess
- Medical University of South Carolina, Charleston, SC
| | - Ryan Jacobs
- Carolinas Medical Center/Levine Cancer Institute, Charlotte, NC
| | | | - Pallawi Torka
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Boyu Hu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Craig Moskowitz
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Supreet Kaur
- University of Texas HSC at San Antonio, San Antonio, TX
| | - Gaurav Goyal
- University of Alabama at Birmingham, Birmingham, AL
| | | | - Andrew Doan
- Children’s Hospital Los Angeles, Los Angeles, CA
| | | | | | | | - Brett Brinker
- Cancer & Hematology Center - W Michigan, Grand Rapids, MI
| | - Namita Sharma
- Geisinger Community Medical Center - Hematology and Oncology, Scranton, PA
| | - Avina Singh
- Fairview Ridges Hospital, Minnesota Oncology, Burnsville, MN
| | - Kristie A. Blum
- Emory University Hospital/Winship Cancer Institute, Atlanta, GA
| | | | | | - David Hodgson
- Princess Margaret Cancer Centre, Toronto, Ontario, CAN
| | | | | | - Anca Prica
- Princess Margaret Cancer Centre, Toronto, Ontario, CAN
| | | | - Richard F. Little
- National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda, MD
| | | | - Michael Crump
- Princess Margaret Cancer Centre, Toronto, Ontario, CAN
| | - Brad Kahl
- Siteman Cancer Center, Washington University, St. Louis, MO
| | | | | | - Joo Y. Song
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Kara M. Kelly
- Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, NY
| | | |
Collapse
|
20
|
Wang H, Wang Z, Zhu S, Li Z, Yang H, Sun P, Zhu M, Zhao X, Shen L, Ou Q, Yang H, Li ZM. Circulating tumor DNA assisting lymphoma genetic feature profiling and identification. Ann Hematol 2024; 103:4135-4144. [PMID: 39012515 DOI: 10.1007/s00277-024-05782-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/24/2024] [Indexed: 07/17/2024]
Abstract
INTRODUCTION Lymphoma tissue biopsies cannot fully capture genetic features due to accessibility and heterogeneity. We aimed to assess the applicability of circulating tumor DNA (ctDNA) for genomic profiling and disease surveillance in classic Hodgkin lymphoma (cHL), primary mediastinal large B-cell lymphoma (PMBCL), and diffuse large B-cell lymphoma (DLBCL). METHODS Tumor tissue and/or liquid biopsies of 49 cHLs, 32 PMBCLs, and 74 DLBCLs were subject to next-generation sequencing targeting 475 genes. The concordance of genetic aberrations in ctDNA and paired tissues was investigated, followed by elevating ctDNA-based mutational landscapes and the correlation between ctDNA dynamics and radiological response/progression. RESULTS ctDNA exhibited high concordance with tissue samples in cHL (78%), PMBCL (84%), and DLBCL (78%). In cHL, more unique mutations were detected in ctDNA than in tissue biopsies (P < 0.01), with higher variant allele frequencies (P < 0.01). Distinct genomic features in cHL, PMBCL, and DLBCL, including STAT6, SOCS1, BTG2, and PIM1 alterations, could be captured by ctDNA alone. Prevalent PD-L1/PD-L2 amplifications were associated with more concomitant alterations in PMBCL (P < 0.01). Moreover, ctDNA fluctuation could reflect treatment responses and indicate relapse before imaging diagnosis. CONCLUSIONS Lymphoma genomic profiling by ctDNA was concordant with that by tumor tissues. ctDNA might also be applied in lymphoma surveillance.
Collapse
Affiliation(s)
- Hongbiao Wang
- Medical Oncology Session No.1, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zhao Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Sujuan Zhu
- Medical Oncology Session No.1, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zhifeng Li
- Medical Oncology Session No.1, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Hang Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Peng Sun
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Minyi Zhu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Xiaotian Zhao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Lu Shen
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Hui Yang
- Department of Hematology, Shunde Hospital, Southern Medical University (the First People's Hospital of Shunde Foshan), No.1 Jiazi Road, Licun, Lunjiao Street, Shunde District, Foshan, 528300, China.
| | - Zhi-Ming Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
21
|
Chen X, Soma L, Murphy C, Tretiakova M, Naresh KN, Fromm JR. Utility of CCR7 to differentiate classic Hodgkin lymphoma and other B-cell lymphomas by flow cytometry and immunohistochemistry. Am J Clin Pathol 2024:aqae119. [PMID: 39288406 DOI: 10.1093/ajcp/aqae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES Classic Hodgkin lymphoma (CHL) is characterized by infrequent neoplastic Hodgkin and Reed-Sternberg (HRS) cells in an inflammatory background. The diagnostic utility of CC-chemokine receptor 7 (CCR7) in CHL was explored using flow cytometry and immunohistochemistry (IHC). METHODS Neoplastic specimens and non-neoplastic lymph nodes were immunophenotyped and CCR7 expression was measured semiquantitatively by flow cytometry (clone 3D12) and IHC (clone 150503). RESULTS Our results showed that CCR7 was expressed on HRS cells in the vast majority of CHL cases (45/48 by flow cytometry, 57/59 by IHC) but rarely expressed in neoplastic cells in diffuse large B-cell lymphoma, not otherwise specified (1/25 by flow cytometry, 2/40 by IHC) and nodular lymphocyte predominant Hodgkin lymphoma (0/4 by flow cytometry, 1/13 by IHC). Primary mediastinal large B-cell lymphoma (PMLBCL) revealed weak CCR7 expression by flow cytometry in most cases (8/10) but only occasionally by IHC (2/12). Both cases (2/2) of T-cell/histiocyte-rich large B-cell lymphoma (THRLBCL) also showed CCR7 expression detected by flow cytometry compared with IHC (0/7). The HRS cells demonstrated a greater percentage of positive cells and greater antigen intensity than the other B-cell lymphomas by IHC. The expression identified by flow cytometry in PMLBCL and THRLBCL but not by IHC suggests that there may be differences in the detection capabilities of the 2 techniques or the 2 CCR7 clones used. CONCLUSIONS The expression of CCR7 in HRS cells suggests its potential utility in differentiating CHL from other B-cell lymphomas. Incorporating CCR7 into flow cytometry and IHC panels may further enhance the diagnostic sensitivity of CHL.
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, US
| | - Lori Soma
- Department of Pathology, City of Hope, Duarte, CA, US
| | - Claire Murphy
- Pathology Consultants, PC, Eugene/Springfield Lab, Springfield, OR, US
| | - Maria Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| | - Kikkeri N Naresh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, US
| | - Jonathan R Fromm
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| |
Collapse
|
22
|
Liu J, Qin J, Liang L, Zhang X, Gao J, Hao Y, Zhao P. Novel insights into the regulation of exosomal PD-L1 in cancer: From generation to clinical application. Eur J Pharmacol 2024; 979:176831. [PMID: 39047964 DOI: 10.1016/j.ejphar.2024.176831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Programmed cell death ligand 1 (PD-L1) interacts with programmed cell death 1 (PD-1), leading to T cell exhaustion and promoting tumor cell survival, ultimately mediating immunosuppression. While FDA-approved monoclonal antibodies targeting the PD-1/PD-L1 interaction have shown success in cancer treatment, some patients experience limited and short-lived therapeutic outcomes. Recent studies have identified PD-L1 expression not only on tumor cell surfaces but also on exosomes, with secretion pathways including both conventional and unconventional endocytosis routes, presenting a unique therapeutic opportunity. Emerging evidence suggests that exosomal PD-L1 contributes to systemic immunosuppression, potentially counteracting the effects of anti-PD-1 checkpoint therapies. However, the significance of exosomal PD-L1 in clinical cancer patients unresponsive to anti-PD-1/PD-L1 immunotherapy, as well as the factors regulating its generation, remain unclear. Moreover, the mechanisms underlying PD-L1 expression on exosomes and its regulation in cancer are yet to be fully elucidated. This review primarily focuses on the mechanisms modulating exosomal PD-L1 generation in cancer, while also outlining its involvement in immunosuppression, tumor proliferation, and response to cancer immunotherapy. Additionally, we explore the potential of exosomal PD-L1 as a cancer biomarker and therapeutic target, aiming to provide a comprehensive overview of this emerging field and its implications for cancer treatment and diagnosis.
Collapse
Affiliation(s)
- Jie Liu
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China; Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Junxia Qin
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Lili Liang
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Xinzhong Zhang
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Jie Gao
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Youwei Hao
- Department of Cardiology, Taiyuan People's Hospital, Taiyuan, 030000, China
| | - Peng Zhao
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China.
| |
Collapse
|
23
|
Spinner MA, Advani RH. Emerging immunotherapies in the Hodgkin lymphoma armamentarium. Expert Opin Emerg Drugs 2024; 29:263-275. [PMID: 38676917 DOI: 10.1080/14728214.2024.2349083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/25/2024] [Indexed: 04/29/2024]
Abstract
INTRODUCTION Brentuximab vedotin and PD-1 inhibitors have improved outcomes for classic Hodgkin lymphoma (cHL), but better therapies are needed for patients who relapse after these agents. Based on an improved understanding of cHL biology, there is a robust pipeline of novel therapies in development. In this review, we highlight emerging immunotherapeutic agents and combinations for cHL. AREAS COVERED We review clinical trials of novel PD-1/PD-L1 inhibitors beyond FDA-approved agents, checkpoint inhibitors targeting CTLA-4, LAG-3, TIM-3, TIGIT, and CD47/SIRPα, PD-1 inhibitor combinations with immunomodulatory agents and epigenetic modifying therapies, antibody-drug conjugates, bispecific antibodies, and cellular therapies including anti-CD30 CAR-T and allogeneic NK cell therapy. We review the key safety and efficacy data from published phase 1-2 studies and highlight trials in progress, including the first phase 3 trial for PD-1 inhibitor-refractory cHL. EXPERT OPINION Many novel immunotherapies hold great promise in cHL. Rational combinations with existing agents and next-generation antibody and CAR-T constructs may improve response rates and durability. Identifying biomarkers of response to these immunotherapies and using more sensitive tools to assess response, such as circulating tumor DNA, may further inform treatment decisions and enable a precision medicine approach in the future.
Collapse
Affiliation(s)
- Michael A Spinner
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Ranjana H Advani
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
24
|
Menéndez V, Solórzano JL, García-Cosío M, Cereceda L, Díaz E, Estévez M, Roncador G, Vega Z, Montalbán C, Kulasinghe A, García JF. Mapping the Spatial Dynamics of the CD4+ T Cell Spectrum in Classical Hodgkin Lymphoma. Mod Pathol 2024; 37:100551. [PMID: 38936478 DOI: 10.1016/j.modpat.2024.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
As around 25% to 30% of classical Hodgkin lymphoma (cHL) patients with advanced stages do not respond to standard therapies, the tumor microenvironment of cHL is one avenue that may be explored with the aim of improving risk stratification. CD4+ T cells are thought to be one of the main cell types in the tumor microenvironment. However, few immune signatures have been studied, and many of these lack related spatial data. Thus, our aim is to spatially resolve the CD4+ T cell subtypes that influence cHL outcome, depicting new immune signatures or transcriptional patterns that are in crosstalk with the tumor cells. This study was conducted using the NanoString GeoMx digital spatial profiling technology, based on the selection of distinct functional areas of patients' tissues followed by gene-expression profiling. The goals were to assess the differences in CD4+ T cell populations between tumor-rich and immune-predominant areas defined by different CD30 and PD-L1 expression levels and seek correlations with clinical metadata. Our results depict a complex map of CD4+ T cells with different functions and differentiation states that are enriched at distinct locations, the flux of cytokines and chemokines that could be related to these, and the specific relationships with the clinical outcome.
Collapse
Affiliation(s)
- Victoria Menéndez
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain
| | - José L Solórzano
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain; Pathology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Mónica García-Cosío
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Laura Cereceda
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain; Pathology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Eva Díaz
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain
| | - Mónica Estévez
- Department of Hematology, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Giovanna Roncador
- Monoclonal Antibodies and Histopathology Units, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Zaira Vega
- Monoclonal Antibodies and Histopathology Units, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Carlos Montalbán
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain
| | - Arutha Kulasinghe
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, Australia
| | - Juan F García
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain; Pathology Department, MD Anderson Cancer Center Madrid, Madrid, Spain.
| |
Collapse
|
25
|
Jeong AR, Trando AH, Thomas SD, Riviere P, Sakowski PJ, Sokol ES, Goodman AM, Kurzrock R. Higher tumor mutational burden and PD-L1 expression correlate with shorter survival in hematologic malignancies. Ther Adv Med Oncol 2024; 16:17588359241273053. [PMID: 39220298 PMCID: PMC11363031 DOI: 10.1177/17588359241273053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/09/2024] [Indexed: 09/04/2024] Open
Abstract
Background The prognostic implications of tumor mutational burden (TMB) and programmed death ligand 1 (PD-L1) expression are poorly studied in hematologic malignancies. Objectives This study aimed to better understand the characteristics and prognostic value of TMB and PD-1/PD-L1 in hematologic malignancies. Design This real-world study was conducted among patients with hematologic malignancies who had next-generation sequencing (NGS) (Foundation Medicine) at the University of California San Diego Moores Cancer Center (2014-2018). Methods TMB was measured by NGS. PD-L1 expression (tumor proportion score, TPS) was measured by immunohistochemistry (classified as high (⩾50%), low (1-49%), and negative (<1%)). Data was curated from the electronic medical records. Results In 388 evaluable patients, the most common diagnoses were B-cell non-Hodgkin lymphoma (NHL) (35%) and Philadelphia chromosome-negative myeloproliferative disorders (16%). Median TMB was 1.6 mutations/Mb (range, 0-46.83). Forty-eight patients (12%) had TMB ⩾10 mutations/Mb, 90% of which were B-cell or T-cell NHL. In 85 samples with available PD-L1 scores, 11 were high; 26, low; and 48, no tumor cell expression. PD-L1 TPS positive (⩾1%) was most common in T-cell NHL (7/9 (77%) cases) followed by B-cell NHL (21/51 (41%) cases). TMB ⩾4 mutations/Mb and PD-L1 score ⩾1% were significantly associated with shorter overall survival (OS) from diagnosis, with hazard ratio (HR) = 1.46 (p = 0.02, 95% confidence interval (CI) 1.05-2.03) and HR = 2.11 (p = 0.04, 95% CI 1.04-4.30), respectively; the relationship was more pronounced when PD-L1 ⩾50% versus <50% was used (HR = 2.80, p = 0.02, 95% CI 1.19-6.59). Higher TMB and higher PD-L1 positivity correlation were significant but weak (Pearson correlation coefficient R 2 = 0.04, p = 0.04). Conclusion TMB ⩾4 mutations/Mb and positive PD-L1 TPS are poor prognostic factors, correlating with shorter OS across hematologic malignancies. Trial registration ClinicalTrials.gov NCT02478931.
Collapse
Affiliation(s)
- Ah-Reum Jeong
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0658, USA
| | - Aaron H. Trando
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sean D. Thomas
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Paul Riviere
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
| | - Patrick J. Sakowski
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Aaron M. Goodman
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Razelle Kurzrock
- Department of Medicine, Medical College of Wisconsin, 8800 West Doyne Avenue, Milwaukee, WI 53226, USA
| |
Collapse
|
26
|
Ghanbari Sevari F, Mehdizadeh A, Abbasi K, Hejazian SS, Raisii M. Cytokine-induced killer cells: new insights for therapy of hematologic malignancies. Stem Cell Res Ther 2024; 15:254. [PMID: 39135188 PMCID: PMC11321231 DOI: 10.1186/s13287-024-03869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Cytokine-induced killer (CIK) cells are a novel subgroup of immune effectors, classified as one of the modified T cell-mediated arms for immunotherapy. These cells exert MHC-unrestricted cytotoxicity against both hematological and solid malignancies with low incidence of treatment-related severe complications. This study reviews the application of CIK cells in treating cases with hematologic malignancies. MAIN BODY CIK cells consist of CD3+/CD56+ natural killer (NK) T cells, CD3-/CD56+ NK cells, and CD3+/CD56- cytotoxic T cells. In this regard, the CD3+/CD56+ NK T cells are the primary effectors. Compared with the previously reported antitumor immune cells, CIK cells are characterized by improved in vitro proliferation and amplification, enhanced migration and invasive capacity to tumor region, more significant antitumor activity, and a broader antitumor spectrum. CIK cells can also induce death in tumor cells via numerous pathways and mechanisms. Hence, CIKs-based therapy has been used in various clinical trials and has shown efficacy with a very low graft versus host disease (GVHD) against several cancers, such as hematologic malignancies, even in relapsing cases, or cases not responding to other therapies. Despite the high content of T cells, CIK cells induce low alloreactivity and, thus, pose a restricted threat of GVHD induction even in MHC-mismatched transplantation cases. Phase 1 and 2 clinical trials of CIK cell therapy have also highlighted satisfactory therapeutic advantages against hematologic cancers, indicating the safety of CIK cells even in haploidentical transplantation settings. CONCLUSION CIK cells have shown promising results in the treatment of hematologic malignancies, especially in combination with other antitumor strategies. However, the existing controversies in achieving desired clinical responses underscore the importance of future studies.
Collapse
Affiliation(s)
- Faezeh Ghanbari Sevari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Abbasi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Sina Hejazian
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mortaza Raisii
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Solórzano JL, Menéndez V, Parra E, Solis L, Salazar R, García-Cosío M, Climent F, Fernández S, Díaz E, Francisco-Cruz A, Khoury J, Jiang M, Tamegnon A, Montalbán C, Melero I, Wistuba I, De Andrea C, F. García J. Multiplex spatial analysis reveals increased CD137 expression and m-MDSC neighboring tumor cells in refractory classical Hodgkin Lymphoma. Oncoimmunology 2024; 13:2388304. [PMID: 39135889 PMCID: PMC11318683 DOI: 10.1080/2162402x.2024.2388304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
The Hodgkin and Reed - Sternberg (HRS) cells in classical Hodgkin Lymphoma (cHL) actively modify the immune tumor microenvironment (TME) attracting immunosuppressive cells and expressing inhibitory molecules. A high frequency of myeloid cells in the TME is correlated with an unfavorable prognosis, but more specific and rare cell populations lack precise markers. Myeloid-derived suppressor cells (MDSCs) have been identified in the peripheral blood of cHL patients, where they appear to be correlated with disease aggressiveness. TNFRSF9 (CD137) is a T cell co-stimulator expressed by monocytic and dendritic cells. Its expression has also been described in HRS cells, where it is thought to play a role in reducing antitumor responses. Here, we perform qualitative and quantitative analyses of lymphocytic and MDSC subtypes and determine the CD137 cell distribution in cHL primary tumors using multiplex immunofluorescence and automated multispectral imaging. The results were correlated with patients' clinical features. Cells were stained with specific panels of immune checkpoint markers (PD-1, PD-L1, CD137), tumor-infiltrating T lymphocytes (CD3, PD-1), and monocytic cells/MDSCs (CD68, CD14, CD33, Arg-1, CD11b). This approach allowed us to identify distinct phenotypes and to analyze spatial interactions between immune subpopulations and tumor cells. The results confirm CD137 expression by T, monocytic and HRS cells. In addition, the expression of CD137, T exhausted cells, and monocytic MDSCs (m-MDSCs) in the vicinity of malignant HRS cells were associated with a worse prognosis. Our findings reveal new elements of the TME that mediate immune escape, and confirm CD137 as a candidate target for immunotherapy in cHL.
Collapse
Affiliation(s)
- José L. Solórzano
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Victoria Menéndez
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Edwin Parra
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Luisa Solis
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | | | | | - Fina Climent
- Pathology Department, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet De Llobregat, Barcelona, Spain
| | - Sara Fernández
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
| | - Eva Díaz
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | | | - Joseph Khoury
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mei Jiang
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Auriole Tamegnon
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Carlos Montalbán
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Ignacio Melero
- University of Navarra and Instituto de Investigación Sanitaria de Navarra, Pamplona, Navarra, Spain
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Carlos De Andrea
- University of Navarra and Instituto de Investigación Sanitaria de Navarra, Pamplona, Navarra, Spain
| | - Juan F. García
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| |
Collapse
|
28
|
Knecht H, Johnson N, Bienz MN, Brousset P, Memeo L, Shifrin Y, Alikhah A, Louis SF, Mai S. Analysis by TeloView ® Technology Predicts the Response of Hodgkin's Lymphoma to First-Line ABVD Therapy. Cancers (Basel) 2024; 16:2816. [PMID: 39199588 PMCID: PMC11352807 DOI: 10.3390/cancers16162816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Classic Hodgkin's lymphoma (cHL) is a curable cancer with a disease-free survival rate of over 10 years. Over 80% of diagnosed patients respond favorably to first-line chemotherapy, but few biomarkers exist that can predict the 15-20% of patients who experience refractory or early relapsed disease. To date, the identification of patients who will not respond to first-line therapy based on disease staging and traditional clinical risk factor analysis is still not possible. Three-dimensional (3D) telomere analysis using the TeloView® software platform has been shown to be a reliable tool to quantify genomic instability and to inform on disease progression and patients' response to therapy in several cancers. It also demonstrated telomere dysfunction in cHL elucidating biological mechanisms related to disease progression. Here, we report 3D telomere analysis on a multicenter cohort of 156 cHL patients. We used the cohort data as a training data set and identified significant 3D telomere parameters suitable to predict individual patient outcomes at the point of diagnosis. Multivariate analysis using logistic regression procedures allowed for developing a predictive scoring model using four 3D telomere parameters as predictors, including the proportion of t-stumps (very short telomeres), which has been a prominent predictor for cHL patient outcome in a previously published study using TeloView® analysis. The percentage of t-stumps was by far the most prominent predictor to identify refractory/relapsing (RR) cHL prior to initiation of adriamycin, bleomycin, vinblastine, and dacarbazine (ABVD) therapy. The model characteristics include an AUC of 0.83 in ROC analysis and a sensitivity and specificity of 0.82 and 0.78 respectively.
Collapse
Affiliation(s)
- Hans Knecht
- Division of Hematology, Jewish General Hospital, McGill University, Montréal, QC H3A 0G4, Canada; (N.J.); (M.N.B.)
| | - Nathalie Johnson
- Division of Hematology, Jewish General Hospital, McGill University, Montréal, QC H3A 0G4, Canada; (N.J.); (M.N.B.)
| | - Marc N. Bienz
- Division of Hematology, Jewish General Hospital, McGill University, Montréal, QC H3A 0G4, Canada; (N.J.); (M.N.B.)
| | - Pierre Brousset
- Toulouse Cancer Center, Université de Toulouse, 31000 Toulouse, France;
| | - Lorenzo Memeo
- Pathology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, 95029 Viagrande, Italy;
| | - Yulia Shifrin
- Telo Genomics Corp., Toronto ON M5G 1L7, Canada; (Y.S.); (A.A.); (S.F.L.)
| | - Asieh Alikhah
- Telo Genomics Corp., Toronto ON M5G 1L7, Canada; (Y.S.); (A.A.); (S.F.L.)
| | - Sherif F. Louis
- Telo Genomics Corp., Toronto ON M5G 1L7, Canada; (Y.S.); (A.A.); (S.F.L.)
| | - Sabine Mai
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3T 2N, Canada;
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
29
|
Ribatti D, Cazzato G, Tamma R, Annese T, Ingravallo G, Specchia G. Immune checkpoint inhibitors targeting PD-1/PD-L1 in the treatment of human lymphomas. Front Oncol 2024; 14:1420920. [PMID: 39091917 PMCID: PMC11291367 DOI: 10.3389/fonc.2024.1420920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Non-Hodgkin lymphomas (NHLs) encompass a diverse group of malignancies arising from B cells, T cells, and natural killer (NK) cells at various stages of differentiation. Conversely, classical Hodgkin lymphomas (cHLs) primarily feature Reed-Sternberg cells (RSCs) amid a background of reactive immune cells. Immunomodulatory pathways, notably the PD-1/PD-L1 axis, play pivotal roles in tumor immune evasion across both NHLs and cHLs. Elevated expression of PD-1 and PD-L1 is observed in a spectrum of lymphomas, influencing prognosis and treatment response. Therapeutically, immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 have revolutionized lymphoma management, particularly in relapsed/refractory cases. Nivolumab and pembrolizumab, among others, have demonstrated efficacy in various B-cell lymphomas, with promising outcomes in cHL. Combination strategies incorporating ICIs with conventional chemotherapy or targeted agents show enhanced efficacy and are being explored extensively. In this review we discuss the most important features of the tumor microenvironment of NHLs and cHLs, address the therapeutic approaches with ICIs and try to outline future perspectives.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy
| | - Gerardo Cazzato
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy
| | - Giuseppe Ingravallo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Medical School, Bari, Italy
| | - Giorgina Specchia
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Medical School, Bari, Italy
| |
Collapse
|
30
|
Zanotta S, Galati D, De Filippi R, Pinto A. Enhancing Dendritic Cell Cancer Vaccination: The Synergy of Immune Checkpoint Inhibitors in Combined Therapies. Int J Mol Sci 2024; 25:7509. [PMID: 39062753 PMCID: PMC11277144 DOI: 10.3390/ijms25147509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 06/27/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Dendritic cell (DC) cancer vaccines are a promising therapeutic approach, leveraging the immune system to fight tumors. These vaccines utilize DCs' ability to present tumor-associated antigens to T cells, triggering a robust immune response. DC vaccine development has progressed through three generations. The first generation involved priming DCs with tumor-associated antigens or messenger RNA outside the body, showing limited clinical success. The second generation improved efficacy by using cytokine mixtures and specialized DC subsets to enhance immunogenicity. The third generation used blood-derived DCs to elicit a stronger immune response. Clinical trials indicate that cancer vaccines have lower toxicity than traditional cytotoxic treatments. However, achieving significant clinical responses with DC immunotherapy remains challenging. Combining DC vaccines with immune checkpoint inhibitors (ICIs), such as anticytotoxic T-lymphocyte Antigen 4 and antiprogrammed death-1 antibodies, has shown promise by enhancing T-cell responses and improving clinical outcomes. These combinations can transform non-inflamed tumors into inflamed ones, boosting ICIs' efficacy. Current research is exploring new checkpoint targets like LAG-3, TIM-3, and TIGIT, considering their potential with DC vaccines. Additionally, engineering T cells with chimeric antigen receptors or T-cell receptors could further augment the antitumor response. This comprehensive strategy aims to enhance cancer immunotherapy, focusing on increased efficacy and improved patient survival rates.
Collapse
Affiliation(s)
- Serena Zanotta
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| | - Domenico Galati
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Antonio Pinto
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| |
Collapse
|
31
|
Zhang W, Zhai Y, Cai Y, Gong X, Jiang Y, Rong R, Zheng C, Zhu B, Zhu HH, Wang H, Li Y, Zhang P. Enhancing immunotherapy efficacy against MHC-I deficient triple-negative breast cancer using LCL161-loaded macrophage membrane-decorated nanoparticles. Acta Pharm Sin B 2024; 14:3218-3231. [PMID: 39027241 PMCID: PMC11252456 DOI: 10.1016/j.apsb.2024.04.009] [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: 12/04/2023] [Revised: 01/21/2024] [Accepted: 04/10/2024] [Indexed: 07/20/2024] Open
Abstract
Current cytotoxic T lymphocyte (CTL) activating immunotherapy requires a major histocompatibility complex I (MHC-I)-mediated presentation of tumor-associated antigens, which malfunctions in around half of patients with triple-negative breast cancer (TNBC). Here, we create a LCL161-loaded macrophage membrane decorated nanoparticle (LMN) for immunotherapy of MHC-I-deficient TNBC. SIRPα on the macrophage membrane helps LMNs recognize CD47-expressing cancer cells for targeted delivery of LCL161, which induces the release of high mobility group protein 1 and proinflammatory cytokines from cancer cells. The released cytokines and high mobility group protein 1 activate antitumor immunity by increasing the intratumoral density of the phagocytic macrophage subtype by 15 times and elevating the intratumoral concentration of CTL lymphotoxin by 4.6 folds. LMNs also block CD47-mediated phagocytosis suppression. LMNs inhibit the growth of MHC-I-deficient TNBC tumors, as well as those resistant to combined therapy of anti-PDL1 antibody and albumin-bound paclitaxel, and prolong the survival of animals, during which process CTLs also play important roles. This macrophage membrane-decorated nanoparticle presents a generalizable platform for increasing macrophage-mediated antitumor immunity for effective immunotherapy of MHC-I-deficient cancers.
Collapse
Affiliation(s)
- Wen Zhang
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yihui Zhai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Gong
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yunxuan Jiang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rong Rong
- Yantai Institute of Materia Medica, Shandong 264000, China
| | - Chao Zheng
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hao Wang
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Yantai Institute of Materia Medica, Shandong 264000, China
| | - Pengcheng Zhang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
32
|
Varma G, Diefenbach C. The role of autologous stem-cell transplantation in classical Hodgkin lymphoma in the modern era. Semin Hematol 2024:S0037-1963(24)00080-5. [PMID: 39039012 DOI: 10.1053/j.seminhematol.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/19/2024] [Accepted: 06/23/2024] [Indexed: 07/24/2024]
Abstract
Despite excellent cure rates with modern front-line regimens, up to 20% of patients with Hodgkin lymphoma will progress through front-line therapy or experience disease relapse. Worldwide, salvage chemotherapy followed by high-dose chemotherapy with autologous stem cell transplantation (HDT/ASCT) is considered the standard of care for these patients and can cure approximately 50% of relapsed or refractory (R/R) patients in the second line. Brentuximab vedotin (BV), an anti-CD30 antibody drug conjugate, and PD1 inhibitors like nivolumab and pembrolizumab, have high response rates in patients who recur after HDT/ASCT. When used prior to HDT/ASCT, BV and PD1 inhibitors appear to dramatically increase the effectiveness of salvage therapies with complete response rates often double those seen with historic chemotherapy-based regimens and durable progression free survival (PFS) post-HDT/ASCT. Emerging data in adults and from pediatric trials showing a durable PFS in a subset of relapsed patients raises the question of whether HDT/ASCT is essential for cure in R/R patients after PD1 based salvage. Future studies will help clarify if ASCT can omitted PD1 based salvage to avoid the potential toxicity of HDT/ASCT without compromising cure.
Collapse
Affiliation(s)
- Gaurav Varma
- Division of Hematology and Medical Oncology, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Division of Hematology and Medical Oncology, NYU Grossman School of Medicine, New York, NY
| | - Catherine Diefenbach
- Division of Hematology and Medical Oncology, Perlmutter Cancer Center at NYU Langone Health, New York, NY; Division of Hematology and Medical Oncology, NYU Grossman School of Medicine, New York, NY.
| |
Collapse
|
33
|
Zak J, Pratumchai I, Marro BS, Marquardt KL, Zavareh RB, Lairson LL, Oldstone MBA, Varner JA, Hegerova L, Cao Q, Farooq U, Kenkre VP, Bachanova V, Teijaro JR. JAK inhibition enhances checkpoint blockade immunotherapy in patients with Hodgkin lymphoma. Science 2024; 384:eade8520. [PMID: 38900864 PMCID: PMC11283877 DOI: 10.1126/science.ade8520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/23/2024] [Indexed: 06/22/2024]
Abstract
Unleashing antitumor T cell activity by checkpoint inhibitor immunotherapy is effective in cancer patients, but clinical responses are limited. Cytokine signaling through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway correlates with checkpoint immunotherapy resistance. We report a phase I clinical trial of the JAK inhibitor ruxolitinib with anti-PD-1 antibody nivolumab in Hodgkin lymphoma patients relapsed or refractory following checkpoint inhibitor immunotherapy. The combination yielded a best overall response rate of 53% (10/19). Ruxolitinib significantly reduced neutrophil-to-lymphocyte ratios and percentages of myeloid suppressor cells but increased numbers of cytokine-producing T cells. Ruxolitinib rescued the function of exhausted T cells and enhanced the efficacy of immune checkpoint blockade in preclinical solid tumor and lymphoma models. This synergy was characterized by a switch from suppressive to immunostimulatory myeloid cells, which enhanced T cell division.
Collapse
Affiliation(s)
- Jaroslav Zak
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | - Isaraphorn Pratumchai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Brett S. Marro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | - Kristi L. Marquardt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | | | - Luke L. Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, USA
| | - Michael B. A. Oldstone
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| | - Judith A. Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, USA
| | - Livia Hegerova
- Division of Hematology, University of Washington School of Medicine, Seattle, USA
| | - Qing Cao
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, USA
| | - Umar Farooq
- Division of Hematology and Oncology and Bone Marrow Transplantation, University of Iowa, Iowa City, USA
| | | | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, USA
| | - John R. Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, USA
| |
Collapse
|
34
|
Zanelli M, Fragliasso V, Parente P, Bisagni A, Sanguedolce F, Zizzo M, Broggi G, Ricci S, Palicelli A, Foroni M, Gozzi F, Gentile P, Morini A, Koufopoulos N, Caltabiano R, Cimino L, Fabozzi M, Cavazza A, Neri A, Ascani S. Programmed Death Ligand 1 (PD-L1) Expression in Lymphomas: State of the Art. Int J Mol Sci 2024; 25:6447. [PMID: 38928153 PMCID: PMC11203507 DOI: 10.3390/ijms25126447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/09/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
The interaction of programmed death-1 (PD-1) on T lymphocytes with its ligands Programmed Death Ligand 1 (PD-L1) and Programmed Death Ligand 2 (PD-L2) on tumor cells and/or tumor-associated macrophages results in inhibitory signals to the T-cell receptor pathway, consequently causing tumor immune escape. PD-L1/PD-L2 are currently used as predictive tissue biomarkers in clinical practice. Virtually PD-L1 levels expressed by tumor cells are associated with a good response to immune checkpoint blockade therapies targeting the PD-1/PD-L1 axis. These therapies restore T-cell antitumor immune response by releasing T-lymphocytes from the inhibitory effects of tumor cells. Immune checkpoint therapies have completely changed the management of patients with solid cancers. This therapeutic strategy is less used in hematological malignancies, although good results have been achieved in some settings, such as refractory/relapsed classic Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Variable results have been obtained in diffuse large B-cell lymphoma and T-cell lymphomas. Immunohistochemistry represents the main technique for assessing PD-L1 expression on tumor cells. This review aims to describe the current knowledge of PD-L1 expression in various types of lymphomas, focusing on the principal mechanisms underlying PD-L1 overexpression, its prognostic significance and practical issues concerning the evaluation of PD-L1 immunohistochemical results in lymphomas.
Collapse
Affiliation(s)
- Magda Zanelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.B.); (S.R.); (A.P.); (M.F.); (A.C.)
| | - Valentina Fragliasso
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Alessandra Bisagni
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.B.); (S.R.); (A.P.); (M.F.); (A.C.)
| | | | - Maurizio Zizzo
- Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (M.Z.); (A.M.); (M.F.)
| | - Giuseppe Broggi
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia” Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (R.C.)
| | - Stefano Ricci
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.B.); (S.R.); (A.P.); (M.F.); (A.C.)
| | - Andrea Palicelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.B.); (S.R.); (A.P.); (M.F.); (A.C.)
| | - Moira Foroni
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.B.); (S.R.); (A.P.); (M.F.); (A.C.)
| | - Fabrizio Gozzi
- Ocular Immunology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.G.); (P.G.); (L.C.)
| | - Pietro Gentile
- Ocular Immunology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.G.); (P.G.); (L.C.)
| | - Andrea Morini
- Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (M.Z.); (A.M.); (M.F.)
| | - Nektarios Koufopoulos
- Second Department of Pathology, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, 15772 Athens, Greece;
| | - Rosario Caltabiano
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia” Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (R.C.)
| | - Luca Cimino
- Ocular Immunology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (F.G.); (P.G.); (L.C.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Massimiliano Fabozzi
- Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (M.Z.); (A.M.); (M.F.)
| | - Alberto Cavazza
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.B.); (S.R.); (A.P.); (M.F.); (A.C.)
| | - Antonino Neri
- Scientific Directorate, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Stefano Ascani
- Pathology Unit, Azienda Ospedaliera Santa Maria di Terni, University of Perugia, 05100 Terni, Italy;
| |
Collapse
|
35
|
Chen F, Sheng J, Li X, Gao Z, Zhao S, Hu L, Chen M, Fei J, Song Z. Unveiling the promise of PD1/PD-L1: A new dawn in immunotherapy for cholangiocarcinoma. Biomed Pharmacother 2024; 175:116659. [PMID: 38692063 DOI: 10.1016/j.biopha.2024.116659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
Cholangiocarcinoma (CCA), a rare yet notably aggressive cancer, has experienced a surge in incidence in recent years. Presently, surgical resection remains the most effective curative strategy for CCA. Nevertheless, a majority of patients with CCA are ineligible for surgical removal at the time of diagnosis. For advanced stages of CCA, the combination of gemcitabine and cisplatin is established as the standard chemotherapy regimen. Despite this, treatment efficacy is often hindered by the development of resistance. In recent times, immune checkpoint inhibitors, particularly those that block programmed death 1 and its ligand (PD1/PD-L1), have emerged as promising strategies against a variety of cancers and are being increasingly integrated into the therapeutic landscape of CCA. A growing body of research supports that the use of PD1/PD-L1 monoclonal antibodies in conjunction with chemotherapy may significantly improve patient outcomes. This article seeks to meticulously review the latest studies on PD1/PD-L1 involvement in CCA, delving into their expression profiles, prognostic significance, contribution to oncogenic processes, and their potential clinical utility.
Collapse
Affiliation(s)
- Fei Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jian Sheng
- Department of Research and Teaching, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoping Li
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhaofeng Gao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Siqi Zhao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingyu Hu
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Minjie Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Jianguo Fei
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
36
|
Ding Y, Jaffe ES. Histopathologic Features and Differential Diagnosis in Challenging Cases of Nodular Lymphocyte Predominant B-cell Lymphoma/Nodular Lymphocyte Predominant Hodgkin Lymphoma. JOURNAL OF CLINICAL AND TRANSLATIONAL PATHOLOGY 2024; 4:61-69. [PMID: 39070246 PMCID: PMC11271245 DOI: 10.14218/jctp.2024.00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Nodular lymphocyte predominant Hodgkin lymphoma was termed "nodular lymphocyte predominant B-cell lymphoma" in the International Consensus Classification (ICC), to emphasize clinical and biological differences from classic Hodgkin lymphoma (CHL). The abbreviation "NLP" represents both terms in the ICC and World Health Organization classifications. Variations in the growth pattern, originally reported as Fan patterns A-F, are designated as either grade 1 or grade 2 in the ICC. NLP is uncommon, and in some cases an accurate diagnosis is challenging. The objectives of this article were to review the histopathologic features of NLP and the differential diagnosis from other key entities including de novo T-cell/histiocyte-rich large B-cell lymphoma (THRLBL) and lymphocyte-rich classic Hodgkin lymphoma (LRCHL). Histologically, NLP Fan pattern E (THRLBL-like) can be indistinguishable from de novo THRLBL. However, focal nodular areas, clustering of tumor cells, presence of few admixed small B-cells or FDC meshworks, and T-cell rosettes favor NLP Fan pattern E and argue against de novo THRLBL. NLP may also be confused with LRCHL. Patients with NLP are younger than those with LRCHL, and LRCHL may show mediastinal involvement. In LRCHL, the nodular pattern often contains eccentrically located small regressed germinal centers and intact small dense FDC meshworks, in contrast to the expanded, and fragmented FDC meshworks in NLP. Neoplastic cells that are positive for CD30 and CD15 but negative for CD20 and CD79a are characteristic of LRCHL. Additionally, Fascin and Gata3 are commonly positive in LRCHL but usually negative in NLP.
Collapse
Affiliation(s)
- Yanna Ding
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Elaine S. Jaffe
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
37
|
Shi H, Sun X, Wu Y, Cui Q, Sun S, Ji N, Liu Y. Targeting the tumor microenvironment in primary central nervous system lymphoma: Implications for prognosis. J Clin Neurosci 2024; 124:36-46. [PMID: 38642434 DOI: 10.1016/j.jocn.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/06/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare extranodal non-Hodgkin lymphoma, and there is limited research on its tumor microenvironment (TME). Nevertheless, more and more studies have evidence that TME has essential effects on tumor cell proliferation, immune escape, and drug resistance. Thus, it is critical to elucidate the role of TME in PCNSL. The understanding of the PCNSL TME is gradually unfolding, including factors that distinguish it from systemic diffuse large B-cell lymphoma (DLBCL). The TME in PCNSL exhibits both transcriptional and spatial intratumor heterogeneity. Cellular interactions between tumor cells and stroma cells reveal immune evasion signaling. The comparative analysis between PCNSL and DLBCL suggests that PCNSL is more likely to be an immunologically deficient tumor. In PCNSL, T cell exhaustion and downregulation of macrophage immune function are accompanied by suppressive microenvironmental factors such as M2 polarized macrophages, endothelin B receptor, HLA depletion, PD-L1, and TIM-3. MMP-9, Integrin-β1, and ICAM-1/LFA-1 play crucial roles in transendothelial migration towards the CNS, while CXCL13/CXCR5, CD44, MAG, and IL-8 are essential for brain parenchymal invasion. Further, macrophages, YKL-40, CD31, CD105, PD-1/PD-L1 axis, osteopontin, galectin-3, aggregative perivascular tumor cells, and HLA deletion may contribute to poor outcomes in patients with PCNSL. This article reviews the effect of various components of TME on the progression and prognosis of PCNSL patients to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Han Shi
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Xuefei Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuchen Wu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Qu Cui
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Shengjun Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Nan Ji
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuanbo Liu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China.
| |
Collapse
|
38
|
Kishida M, Fujisawa M, Steidl C. Molecular biomarkers in classic Hodgkin lymphoma. Semin Hematol 2024:S0037-1963(24)00069-6. [PMID: 38969539 DOI: 10.1053/j.seminhematol.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/27/2024] [Indexed: 07/07/2024]
Abstract
Classic Hodgkin lymphoma is a unique B-cell derived malignancy featuring rare malignant Hodgkin and Reed Sternberg (HRS) cells that are embedded in a quantitively dominant tumor microenvironment (TME). Treatment of classic Hodgkin lymphoma has significantly evolved in the past decade with improving treatment outcomes for newly diagnosed patients and the minority of patients suffering from disease progression. However, the burden of toxicity and treatment-related long-term sequelae remains high in a typically young patient population. This highlights the need for better molecular biomarkers aiding in risk-adapted treatment strategies and predicting response to an increasing number of available treatments that now prominently involve multiple immunotherapy options. Here, we review modern molecular biomarker approaches that reflect both the biology of the malignant HRS cells and cellular components in the TME, while holding the promise to improve diagnostic frameworks for clinical decision-making and be feasible in clinical trials and routine practice. In particular, technical advances in sequencing and analytic pipelines using liquid biopsies, as well as deep phenotypic characterization of tissue architecture at single-cell resolution, have emerged as the new frontier of biomarker development awaiting further validation and implementation in routine diagnostic procedures.
Collapse
Affiliation(s)
- Makoto Kishida
- Centre for Lymphoid Cancer department, BC Cancer, Vancouver, British Columbia, Canada
| | - Manabu Fujisawa
- Centre for Lymphoid Cancer department, BC Cancer, Vancouver, British Columbia, Canada; Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Christian Steidl
- Centre for Lymphoid Cancer department, BC Cancer, Vancouver, British Columbia, Canada; Institute of Medicine, University of Tsukuba, Ibaraki, Japan; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
39
|
Li K, Nie H, Jin R, Wu X. Mesenchymal stem cells-macrophages crosstalk and myeloid malignancy. Front Immunol 2024; 15:1397005. [PMID: 38779660 PMCID: PMC11109455 DOI: 10.3389/fimmu.2024.1397005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
As major components of the tumor microenvironment, both mesenchymal stem cells (MSCs) and macrophages can be remodelled and exhibit different phenotypes and functions during tumor initiation and progression. In recent years, increasing evidence has shown that tumor-associated macrophages (TAMs) play a crucial role in the growth, metastasis, and chemotherapy resistance of hematological malignancies, and are associated with poor prognosis. Consequently, TAMs have emerged as promising therapeutic targets. Notably, MSCs exert a profound influence on modulating immune cell functions such as macrophages and granulocytes, thereby playing a crucial role in shaping the immunosuppressive microenvironment surrounding tumors. However, in hematological malignancies, the cellular and molecular mechanisms underlying the interaction between MSCs and macrophages have not been clearly elucidated. In this review, we provide an overview of the role of TAMs in various common hematological malignancies, and discuss the latest advances in understanding the interaction between MSCs and macrophages in disease progression. Additionally, potential therapeutic approaches targeting this relationship are outlined.
Collapse
Affiliation(s)
- Kun Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Nie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Kosydar S, Ansell SM. The biology of classical Hodgkin lymphoma. Semin Hematol 2024:S0037-1963(24)00059-3. [PMID: 38824068 DOI: 10.1053/j.seminhematol.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/05/2024] [Indexed: 06/03/2024]
Abstract
Classical Hodgkin lymphoma (cHL) is distinguished by several important biological characteristics. The presence of Hodgkin Reed Sternberg (HRS) cells is a defining feature of this disease. The tumor microenvironment with relatively few HRS cells in an expansive infiltrate of immune cells is another key feature. Numerous cell-cell mediated interactions and a plethora of cytokines in the tumor microenvironment collectively work to promote HRS cell growth and survival. Aberrancy and constitutive activation of core signal transduction pathways are a hallmark trait of cHL. Genetic lesions contribute to these dysregulated pathways and evasion of the immune system through a variety of mechanisms is another notable feature of cHL. While substantial elucidation of the biology of cHL has enabled advancements in therapy, increased understanding in the future of additional mechanisms driving cHL may lead to new treatment opportunities.
Collapse
Affiliation(s)
| | - Stephen M Ansell
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
41
|
Milrod CJ, Pelcovits A, Ollila TA. Immune checkpoint inhibitors in advanced and relapsed/refractory Hodgkin lymphoma: current applications and future prospects. Front Oncol 2024; 14:1397053. [PMID: 38699638 PMCID: PMC11063339 DOI: 10.3389/fonc.2024.1397053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Classic Hodgkin lymphoma (cHL) treatment paradigms are undergoing a shift with the integration of immune checkpoint inhibitors (ICIs) into both first-line and relapsed/refractory (R/R) regimens. In first-line therapy, the synergy between ICIs and chemotherapy may surpass the previous standards of ABVD and BV-AVD established by landmark trials including RATHL and ECHELON-1. In R/R disease, the combination of ICIs with chemotherapy has begun to challenge the paradigm of chemotherapy as a bridge to consolidative autologous stem cell transplantation. The clinical advances heralded by ICI offer unique challenges to management. ICI treatment and the associated inflammatory response can make the traditional timing and modalities of treatment response assessment difficult to interpret. In contrast to ABVD and BV-AVD, pembrolizumab-AVD results in PET2 positivity rates that are higher and less predictive of treatment response even when ultimate outcomes may be superior. This suggests that the predictive value of PET2 may be less reliable in the ICI era, prompting a reevaluation of response assessment strategies. Looking forward, circulating tumor DNA (ctDNA) may be a promising tool in response-adapted therapy. Its potential to complement or even supersede PET scans in predicting response to ICIs represents a critical advancement. The integration of ctDNA analysis holds the promise of refining response-adapted approaches and enhancing precision in therapeutic decision-making for patients with cHL. This review navigates the evolving landscape of cHL therapy, emphasizing the paradigmatic shift brought about by ICIs. This article explores the impact of combining ICIs with chemotherapy in both relapsed/refractory and first-line settings, scrutinizes the challenges posed to response-adapted therapy by ICIs, and highlights the potential role of ctDNA as an adjunct in refining response-adapted strategies for cHL.
Collapse
|
42
|
Medeiros LJ, Chadburn A, Natkunam Y, Naresh KN. Fifth Edition of the World Health Classification of Tumors of the Hematopoietic and Lymphoid Tissues: B-cell Neoplasms. Mod Pathol 2024; 37:100441. [PMID: 38309432 DOI: 10.1016/j.modpat.2024.100441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
We review B-cell neoplasms in the 5th edition of the World Health Organization classification of hematolymphoid tumors (WHO-HEM5). The revised classification is based on a multidisciplinary approach including input from pathologists, clinicians, and other experts. The WHO-HEM5 follows a hierarchical structure allowing the use of family (class)-level definitions when defining diagnostic criteria are partially met or a complete investigational workup is not possible. Disease types and subtypes have expanded compared with the WHO revised 4th edition (WHO-HEM4R), mainly because of the expansion in genomic knowledge of these diseases. In this review, we focus on highlighting changes and updates in the classification of B-cell lymphomas, providing a comparison with WHO-HEM4R, and offering guidance on how the new classification can be applied to the diagnosis of B-cell lymphomas in routine practice.
Collapse
Affiliation(s)
- L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Amy Chadburn
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Kikkeri N Naresh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle; Section of Pathology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle
| |
Collapse
|
43
|
Okatani T, Nishimura MF, Egusa Y, Yoshida S, Nishimura Y, Nishikori A, Yoshino T, Yamamoto H, Sato Y. Analysis of Notch1 protein expression in methotrexate-associated lymphoproliferative disorders. J Clin Exp Hematop 2024; 64:1-9. [PMID: 38281745 PMCID: PMC11079991 DOI: 10.3960/jslrt.23038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 01/30/2024] Open
Abstract
Methotrexate (MTX)-associated lymphoproliferative disorder (MTX-LPD) is a lymphoproliferative disorder in patients treated with MTX. The mechanism of pathogenesis is still elusive, but it is thought to be a complex interplay of factors, such as underlying autoimmune disease activity, MTX use, Epstein-Barr virus infection, and aging. The NOTCH genes encode receptors for a signaling pathway that regulates various fundamental cellular processes, such as proliferation and differentiation during embryonic development. Mutations of NOTCH1 have been reported in B-cell tumors, including chronic lymphocytic leukemia/lymphoma, mantle cell lymphoma, and diffuse large B-cell lymphoma (DLBCL). Recently, it has also been reported that NOTCH1 mutations are found in post-transplant lymphoproliferative disorders, and in CD20-positive cells in angioimmunoblastic T-cell lymphoma, which might be associated with lymphomagenesis in immunodeficiency. In this study, to investigate the association of NOTCH1 in the pathogenesis of MTX-LPD, we evaluated protein expression of Notch1 in nuclei immunohistochemically in MTX-LPD cases [histologically DLBCL-type (n = 24) and classical Hodgkin lymphoma (CHL)-type (n = 24)] and de novo lymphoma cases [DLBCL (n = 19) and CHL (n = 15)]. The results showed that among MTX-LPD cases, the expression of Notch1 protein was significantly higher in the DLBCL type than in the CHL type (P < 0.001). In addition, among DLBCL morphology cases, expression of Notch1 tended to be higher in MTX-LPD than in the de novo group; however this difference was not significant (P = 0.0605). The results showed that NOTCH1 may be involved in the proliferation and tumorigenesis of B cells under the use of MTX. Further research, including genetic studies, is necessary.
Collapse
|
44
|
Ohsawa K, Momose S, Nishikori A, Nishimura MF, Gion Y, Sawada K, Higashi M, Tokuhira M, Tamaru JI, Sato Y. Copy Number Analysis of 9p24.1 in Classic Hodgkin Lymphoma Arising in Immune Deficiency/Dysregulation. Cancers (Basel) 2024; 16:1298. [PMID: 38610976 PMCID: PMC11011107 DOI: 10.3390/cancers16071298] [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: 02/12/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024] Open
Abstract
A subset of patients with rheumatoid arthritis receiving methotrexate develop immune deficiencies and dysregulation-associated lymphoproliferative disorders. Patients with these disorders often exhibit spontaneous regression after MTX withdrawal; however, chemotherapeutic intervention is frequently required in patients with classic Hodgkin lymphoma arising in immune deficiency/dysregulation. In this study, we examined PD-L1 expression levels and 9p24.1 copy number alterations in 27 patients with classic Hodgkin lymphoma arising from immune deficiency/dysregulation. All patients demonstrated PD-L1 protein expression and harbored 9p24.1 copy number alterations on the tumor cells. When comparing clinicopathological data and associations with 9p24.1 copy number features, the copy gain group showed a significantly higher incidence of extranodal lesions and clinical stages than the amplification group. Notably, all cases in the amplification group had latency type II, while 6/8 (75%) in the copy gain group had latency type II, and 2/8 (25%) had latency type I. Thus, a subset of the copy-gain group demonstrated more extensive extranodal lesions and higher clinical stages. This finding speculates the presence of a genetically distinct subgroup within the group of patients who develop immune deficiencies and dysregulation-associated lymphoproliferative disorders, which may explain certain characteristic features.
Collapse
Affiliation(s)
- Kumiko Ohsawa
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama 700-8558, Japan; (K.O.); (A.N.); (M.F.N.)
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan; (S.M.); (K.S.); (M.H.); (J.-i.T.)
| | - Shuji Momose
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan; (S.M.); (K.S.); (M.H.); (J.-i.T.)
| | - Asami Nishikori
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama 700-8558, Japan; (K.O.); (A.N.); (M.F.N.)
| | - Midori Filiz Nishimura
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama 700-8558, Japan; (K.O.); (A.N.); (M.F.N.)
| | - Yuka Gion
- Department of Medical Technology, Faculty of Health Sciences, Ehime Prefectural University of Health Sciences, Tobe 791-2101, Japan;
| | - Keisuke Sawada
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan; (S.M.); (K.S.); (M.H.); (J.-i.T.)
| | - Morihiro Higashi
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan; (S.M.); (K.S.); (M.H.); (J.-i.T.)
| | - Michihide Tokuhira
- Department of Hematology, Japan Community Health Care Organization Saitama Medical Center, Saitama 330-0074, Japan
| | - Jun-ichi Tamaru
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan; (S.M.); (K.S.); (M.H.); (J.-i.T.)
- PCL Japan, Pathology and Cytology Center, Saitama 331-9530, Japan
| | - Yasuharu Sato
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama 700-8558, Japan; (K.O.); (A.N.); (M.F.N.)
| |
Collapse
|
45
|
Aoki T, Jiang A, Xu A, Yin Y, Gamboa A, Milne K, Takata K, Miyata-Takata T, Chung S, Rai S, Wu S, Warren M, Strong C, Goodyear T, Morris K, Chong LC, Hav M, Colombo AR, Telenius A, Boyle M, Ben-Neriah S, Power M, Gerrie AS, Weng AP, Karsan A, Roth A, Farinha P, Scott DW, Savage KJ, Nelson BH, Merchant A, Steidl C. Spatially Resolved Tumor Microenvironment Predicts Treatment Outcomes in Relapsed/Refractory Hodgkin Lymphoma. J Clin Oncol 2024; 42:1077-1087. [PMID: 38113419 PMCID: PMC10950131 DOI: 10.1200/jco.23.01115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/12/2023] [Accepted: 10/04/2023] [Indexed: 12/21/2023] Open
Abstract
PURPOSE About a third of patients with relapsed or refractory classic Hodgkin lymphoma (r/r CHL) succumb to their disease after high-dose chemotherapy followed by autologous stem-cell transplantation (HDC/ASCT). Here, we aimed to describe spatially resolved tumor microenvironment (TME) ecosystems to establish novel biomarkers associated with treatment failure in r/r CHL. PATIENTS AND METHODS We performed imaging mass cytometry (IMC) on 71 paired primary diagnostic and relapse biopsies using a marker panel specific to CHL biology. For each cell type in the TME, we calculated a spatial score measuring the distance of nearest neighbor cells to the malignant Hodgkin Reed Sternberg cells within the close interaction range. Spatial scores were used as features in prognostic model development for post-ASCT outcomes. RESULTS Highly multiplexed IMC data revealed shared TME patterns in paired diagnostic and early r/r CHL samples, whereas TME patterns were more divergent in pairs of diagnostic and late relapse samples. Integrated analysis of IMC and single-cell RNA sequencing data identified unique architecture defined by CXCR5+ Hodgkin and Reed Sternberg (HRS) cells and their strong spatial relationship with CXCL13+ macrophages in the TME. We developed a prognostic assay (RHL4S) using four spatially resolved parameters, CXCR5+ HRS cells, PD1+CD4+ T cells, CD68+ tumor-associated macrophages, and CXCR5+ B cells, which effectively separated patients into high-risk versus low-risk groups with significantly different post-ASCT outcomes. The RHL4S assay was validated in an independent r/r CHL cohort using a multicolor immunofluorescence assay. CONCLUSION We identified the interaction of CXCR5+ HRS cells with ligand-expressing CXCL13+ macrophages as a prominent crosstalk axis in relapsed CHL. Harnessing this TME biology, we developed a novel prognostic model applicable to r/r CHL biopsies, RHL4S, opening new avenues for spatial biomarker development.
Collapse
Affiliation(s)
- Tomohiro Aoki
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Princess Margaret Cancer Centre—University Health Network, Toronto, Ontario, Canada
| | - Aixiang Jiang
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Yifan Yin
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | | | - Katy Milne
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Katsuyoshi Takata
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
- Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Shanee Chung
- Leukemia/Bone Marrow Transplant Program of BC, BC Cancer, Vancouver, British Columbia, Canada
| | - Shinya Rai
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | - Shaocheng Wu
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Mary Warren
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Celia Strong
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Talia Goodyear
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Kayleigh Morris
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Lauren C. Chong
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | | | | | - Adele Telenius
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | - Merrill Boyle
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | - Susana Ben-Neriah
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | - Maryse Power
- Leukemia/Bone Marrow Transplant Program of BC, BC Cancer, Vancouver, British Columbia, Canada
| | - Alina S. Gerrie
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | - Andrew P. Weng
- Terry Fox Laboratory, BC Cancer, Vancouver, British Columbia, Canada
| | - Aly Karsan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew Roth
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Pedro Farinha
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - David W. Scott
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | - Kerry J. Savage
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
| | - Brad H. Nelson
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | | | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
46
|
Bednarska K, Thillaiyampalam G, Mujaj S, Nourse J, Gunawardana J, Sabdia MB, Law SC, Pilaar A, Cui Q, de Long LM, Vari F, Gandhi MK, Cristino AS. The IRE1α Endonuclease Plays a Dual Role in Regulating the XBP1/miRNA-34a Axis and PD-1 Expression within Natural Killer Cells in Hodgkin Lymphoma. Acta Haematol 2024; 147:676-691. [PMID: 38479365 DOI: 10.1159/000536044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 12/26/2023] [Indexed: 12/05/2024]
Abstract
INTRODUCTION Hodgkin lymphoma (HL) is deficient in major histocompatibility complex class I, rendering it susceptible to antitumoral immunity by natural killer (NK) cells. Despite the functional impairment of PD-1+ NK cells in HL, the underlying mechanisms of NK cell dysfunction remain unclear. METHODS This study involved 14 HL patients and SNK10/KHYG-1 cell lines to assess NK cell activation against cancer cells. Activation was measured through transcript (PCR) and protein expression (flow cytometry). Regulatory mechanisms associated with IRE1α activation were validated through knockdown and luciferase reporter assays. RESULTS Our findings reveal a novel role for IRE1α-endonuclease in fine-tuning NK cell effector functions by orchestrating the XBP1s/microRNA-34a-5p/PD-1 axis. When NK cells encounter cancer cells, IRE1α endonuclease activates the decay of microRNA-34a-5p, resulting in increased expression of XBP1s and PD-1. IRE1α-endonuclease activation enhances NK cell functions while promoting PD-1 expression. In turn, PD-1 is directly regulated by microRNA-34a-5p, which binds to the 3'UTR of PD-1 transcript to repress PD-1 protein on the NK cell surface. Importantly, IRE1α-pathway activation is impaired in NK cells from HL patients. CONCLUSION The IRE1α endonuclease emerges as a key player, simultaneously regulating the XBP1s/microRNA-34a-5p/PD-1 axis in NK cells, a process disrupted in HL. Targeting the IRE1α-pathway holds promise as a therapeutic strategy to optimize NK cell functions in Hodgkin lymphoma treatments.
Collapse
Affiliation(s)
- Karolina Bednarska
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Gayathri Thillaiyampalam
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Sally Mujaj
- Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Jamie Nourse
- Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Jay Gunawardana
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Muhammed B Sabdia
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Soi C Law
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Anna Pilaar
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Qingyan Cui
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Lilia M de Long
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Frank Vari
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Maher K Gandhi
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
- Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Alexandre S Cristino
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
47
|
Yeo YY, Qiu H, Bai Y, Zhu B, Chang Y, Yeung J, Michel HA, Wright K, Shaban M, Sadigh S, Nkosi D, Shanmugam V, Rock P, Tung Yiu SP, Cramer P, Paczkowska J, Stephan P, Liao G, Huang AY, Wang H, Chen H, Frauenfeld L, Mitra B, Gewurz BE, Schürch CM, Zhao B, Nolan GP, Zhang B, Shalek AK, Angelo M, Mahmood F, Ma Q, Burack WR, Shipp MA, Rodig SJ, Jiang S. Epstein-Barr Virus Orchestrates Spatial Reorganization and Immunomodulation within the Classic Hodgkin Lymphoma Tumor Microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583586. [PMID: 38496566 PMCID: PMC10942289 DOI: 10.1101/2024.03.05.583586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Classic Hodgkin Lymphoma (cHL) is a tumor composed of rare malignant Hodgkin and Reed-Sternberg (HRS) cells nested within a T-cell rich inflammatory immune infiltrate. cHL is associated with Epstein-Barr Virus (EBV) in 25% of cases. The specific contributions of EBV to the pathogenesis of cHL remain largely unknown, in part due to technical barriers in dissecting the tumor microenvironment (TME) in high detail. Herein, we applied multiplexed ion beam imaging (MIBI) spatial pro-teomics on 6 EBV-positive and 14 EBV-negative cHL samples. We identify key TME features that distinguish between EBV-positive and EBV-negative cHL, including the relative predominance of memory CD8 T cells and increased T-cell dysfunction as a function of spatial proximity to HRS cells. Building upon a larger multi-institutional cohort of 22 EBV-positive and 24 EBV-negative cHL samples, we orthogonally validated our findings through a spatial multi-omics approach, coupling whole transcriptome capture with antibody-defined cell types for tu-mor and T-cell populations within the cHL TME. We delineate contrasting transcriptomic immunological signatures between EBV-positive and EBV-negative cases that differently impact HRS cell proliferation, tumor-immune interactions, and mecha-nisms of T-cell dysregulation and dysfunction. Our multi-modal framework enabled a comprehensive dissection of EBV-linked reorganization and immune evasion within the cHL TME, and highlighted the need to elucidate the cellular and molecular fac-tors of virus-associated tumors, with potential for targeted therapeutic strategies.
Collapse
|
48
|
Sun P, Yang H, Wang Y, Zhao B, Nie M, Huang K, Li Z. Tislelizumab monotherapy in patients with previously untreated early-stage classical Hodgkin lymphoma: a real-world study. Ann Hematol 2024; 103:793-801. [PMID: 37953379 DOI: 10.1007/s00277-023-05541-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
The anti-PD-1 antibodies have been reported to show a striking effect in relapsed and refractory(R/R) classical Hodgkin lymphoma (cHL), however, there is still limited real-world data assessing the role of anti-PD-1 antibody monotherapy in early-stage cHL. In this retrospective analysis, we reported the effectiveness and safety of tislelizumab monotherapy in the first-line therapy of early-stage cHL. Twenty-three consecutive patients (10 males and 13 females) with previously untreated stage I A-II B cHL were included. At interim evaluation after 2 doses of tislelizumab monotherapy, 11 of 23 patients (47.8%) achieved complete response (CR). At the end of tislelizumab monotherapy (EOTM), objective response was observed in 22 of 23 patients (95.7%), with CR in 16 patients (69.6%). Among six patients with PR-EOTM, two patients underwent 4 cycles of ABVD chemotherapy and one patient underwent 4 cycles of tislelizumab plus AVD. One patient who developed progressive disease (PD) after 4 doses of tislelizumab subsequently underwent 4 cycles of ABVD chemotherapy. Except for four patients with CR-EOTM, consolidative radiotherapy was given to 19 patients. All patients obtained CR at the end of all treatments. With a median follow-up time of 21.3 months (range, 6.9-32.7 months), the estimated 2-year PFS rate and 2-year OS rate were 95.65% and 100%, respectively. Except for grade 3 lymphocyte count decreased, no other grade 3/4 TRAE was observed. In addition, no serious AE was reported. Our preliminary data observed that tislelizumab monotherapy was safe and highly effective in previously untreated early-stage cHL.
Collapse
Affiliation(s)
- Peng Sun
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Hang Yang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Yu Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Baitian Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
- Department of Clinical Trials Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Man Nie
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Kangming Huang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Zhiming Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China.
| |
Collapse
|
49
|
Zhang Q, Zhu B, Yang H, Li F, Qu Y, Lu L, Zhang Q. Exploration of YBX1 role in the prognostic value and immune characteristics by single-cell and bulk sequencing analysis for liver hepatocellular carcinoma. J Gene Med 2024; 26:e3680. [PMID: 38448368 DOI: 10.1002/jgm.3680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Y-box binding protein 1 (YBX1) plays a variety of roles in progression of multiple tumors. However, the role of YBX1 in prognostic value and immune regulation for liver hepatocellular carcinoma (LIHC) remains unclear. The present study aimed to examine the effect of YBX1 on the regulation of tumor immunity and survival prediction in LIHC patients. METHODS YBX1-related expression profiles and single-cell and bulk sequencing analysis were performed using online databases. YBX1 expression was validated by a quantitative real-time PCR (qRT-PCR), western blotting and immunohistochemistry. Univariate/multivariate Cox regression analysis was performed to determine independent predictors of overall survival (OS). The ESTIMATE (i.e., Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data) algorithm and Tumor Immune Dysfunction and Exclusion (TIDE) analysis were used to assess the relationships between YBX1 and LIHC immunity. RESULTS YBX1 was over-expressed in LIHC tissues and cell lines. High YBX1 expression was significantly associated with poor OS. Univariate/multivariate Cox regression analysis revealed that YBX1 was an independent prognostic factor for LIHC. Gene set enrichment analysis revealed that YBX1 was associated with multiple signaling pathways correlated to LIHC. Additionally, YBX1 was expressed in multiple immune cells and was significantly correlated with immune cells, immune checkpoint markers and tumor immune microenvironment. The TIDE analysis demonstrated that LIHC patients with high YBX1 expression showed a higher T-cell dysfunction score and a higher exclusion score, as well as poorer immunotherapy response. CONCLUSIONS YBX1 plays crucial oncogenic roles in LIHC and is closely associated with the immune defense system. YBX1 inhibition may serve as a potential treatment for LIHC.
Collapse
Affiliation(s)
- Qingqing Zhang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingye Zhu
- Department of Urology, Affiliated Nantong Hospital of Shanghai University, The Sixth People's Hospital of Nantong, Nantong, Jiangsu Province, China
| | - Hongyan Yang
- Nursing Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Qu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lungen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Zhang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Libert D, Zhao S, Younes S, Mosquera AP, Bharadwaj S, Ferreira C, Natkunam Y. TIGIT is Frequently Expressed in the Tumor Microenvironment of Select Lymphomas: Implications for Targeted Therapy. Am J Surg Pathol 2024; 48:337-352. [PMID: 38148663 PMCID: PMC10876169 DOI: 10.1097/pas.0000000000002168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Immune checkpoint inhibitors against Programmed Cell Death Protein 1/Programmed Cell (PD-1/PD-L1) and CTLA-4/B7 axes have had limited success in hematologic malignancies, requiring the need to explore alternative targets such as T-cell immunoreceptor with Ig and ITIM domains (TIGIT)/CD155 to improve durable clinical responses. We undertook this study to investigate the expression profile of TIGIT such that the potential efficacy of TIGIT blockade could be mapped among lymphoma subtypes. We validated an immunohistochemical assay for TIGIT and evaluated its expression in lymphoma and tumor microenvironment (TME) cells in 661 lymphoma/leukemia biopsies. Multiplex immunofluorescence was used for correlation with normal TME cell subsets. Tumor or TME TIGIT-positivity was defined as moderate to strong membrane staining in at least 10% of tumor or TME cells, respectively. TME TIGIT expression was correlated with overall survival and progression-free survival and comparison with PD-L1 expression. In most cases, lymphoma cells were TIGIT-negative except for angioimmunoblastic and peripheral T-cell lymphomas, which showed 91% and 47% positivity, respectively. A high proportion of small B-cell lymphoma and anaplastic large cell lymphoma cases had TIGIT-positive TME cells. Chronic lymphocytic leukemia/small lymphocytic lymphoma patients with TIGIT-negative TME cells showed significantly shorter overall survival ( P =0.04). No other statistically significant differences were found. When TIGIT was expressed in TME cells, there were a comparable number of TIGIT-positive only and dual TIGIT/PD-L1 positive cases except for more TIGIT-positive only cases in CLL/SLL. TIGIT expression shows distinctive profiles among lymphoma subtypes. Chronic lymphocytic leukemia/small lymphocytic lymphoma and anaplastic large cell lymphoma demonstrated high TME TIGIT expression compared with PD-L1, with a high proportion of dual TIGIT and PD-L1-positivity. Our results are likely to contribute to the design and correlative study of therapeutic response in clinical trials targeting TIGIT alone or in combination with PD1/PDL1.
Collapse
|