1
|
Courtois A, Allaume P, Raby M, Pastoret C, Droitcourt C, Le Naourès C, Adamski H, Dupuy A, Le Gall F, Kammerer-Jacquet SF. Differential Expression of p53 in Mycosis Fungoides, Sezary Syndromes, and Their Transformed Forms. Am J Dermatopathol 2024:00000372-990000000-00471. [PMID: 39660957 DOI: 10.1097/dad.0000000000002898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
ABSTRACT Mycosis fungoides (MF) and Sezary syndrome (SS) are common entities among primary cutaneous lymphomas. Large cell transformation is challenging for diagnosis and therapy. Molecular mechanisms by which these lymphomas undergo this transformation are poorly defined. We studied the immunohistochemical status of p53 in these entities and assessed whether p53 expression could be a useful tool for diagnosis and assessment of transformation. We extracted patients with transformed and untransformed SS or MF from the French Study Group on Cutaneous Lymphoma database between 2014 and 2021, followed in the Rennes University Hospital. An immunohistochemical study of p53 expression was performed on the biopsies sampled as part of routine care. We compared p53 overexpression in the different groups. We included 25 patients with MF, 7 patients with transformed MF (T-MF), 11 patients with SS, and 5 patients with transformed SS (T-SS). Using a cut-off set at 30% expression of neoplastic cells, we noted an overexpression of p53 in T-MF and T-SS compared with nontransformed forms (47% vs. 12%, respectively, P < 0.01) and in MF compared with SS (23% vs. 7%, respectively, P < 0.01). Overexpression of p53 with a cut-off at 30% therefore seems to be a discriminating tool in the differential diagnosis of MF/SS versus their transformed forms as well as the differential diagnosis between MF and SS.
Collapse
Affiliation(s)
- Anna Courtois
- Department of Pathology, Rennes University Hospital, France
| | - Pierre Allaume
- Department of Pathology, Rennes University Hospital, France
| | - Maxime Raby
- Department of Dermatology, Rennes University Hospital, France
| | - Cédric Pastoret
- Department of Hematology, Rennes University Hospital, France; and
| | | | | | - Henri Adamski
- Department of Dermatology, Rennes University Hospital, France
| | - Alain Dupuy
- Department of Dermatology, Rennes University Hospital, France
| | | | | |
Collapse
|
2
|
Purnak S, Hosing C, Dabaja B, Bassett RL, Huen A, Duvic M. On the Way to Curing Advanced-Stage Mycosis Fungoides/Sézary Syndrome. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:827-836. [PMID: 39107202 DOI: 10.1016/j.clml.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 08/09/2024]
Abstract
INTRODUCTION/BACKGROUND Advanced-stage mycosis fungoides (MF) and Sézary syndrome (SS) have poor prognosis with median survivals of less than 5 years. Although a variety of treatments are approved for MF/SS patients, durable complete remissions (CR) are rare. PATIENTS AND METHODS Advanced-stage MF or SS patients who achieved CR and maintained in CR or stage IA for more than 10 years were identified by a retrospective search of the principal investigator's database. RESULTS Of 2266 patients diagnosed with MF or SS, 23 patients with advanced-stage MF/SS (6 IIB, 1 IIIB, 5 IVA1, 3 IVA2, 8 IVB) who achieved CR and maintained in CR or stage IA for ≥ 10 years were identified. As final/curative treatment, 11 patients underwent allogeneic stem cell transplantation (SCT). Most patients presented at young age, underwent SCT with reduced intensity conditioning regimen, had matched related donors, and controllable post-transplant graft versus host disease. Eleven patients were treated with TSEB as part of combined modality protocol in 2 patients and debulking therapy before allogeneic SCT in 9 patients. Five stage IIB patients achieved CR with radiotherapy. Four patients with blood involvement were treated with extracorporeal photopheresis (ECP) in combination with long-term antibiotics and immunomodulatory agents. Long-term antibiotics were given to 14 patients. CONCLUSION TSEB followed by allogeneic SCT, radiotherapy, ECP plus long-term antibiotics and immunomodulatory agents were the most common curative/final treatments found in our patients. We are reporting the details of our long-term complete responders' treatment course in the hopes of obtaining more cure responses in the future.
Collapse
Affiliation(s)
- Seda Purnak
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Chitra Hosing
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bouthaina Dabaja
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Roland L Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Auris Huen
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Madeleine Duvic
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
3
|
Gao J, Liu J, Lu J, Zhang X, Zhang W, Li Q, Cai J, Li M, Gan Y, Tang Y, Wu S. SKAP1 Expression in Cancer Cells Enhances Colon Tumor Growth and Impairs Cytotoxic Immunity by Promoting Neutrophil Extracellular Trap Formation via the NFATc1/CXCL8 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403430. [PMID: 39269257 PMCID: PMC11538704 DOI: 10.1002/advs.202403430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/15/2024] [Indexed: 09/15/2024]
Abstract
The mechanisms underlying the development and progression of colon cancer are not fully understood. Herein, Src kinase associated phosphoprotein 1 (SKAP1), an immune cell adaptor, is identified as a novel colon cancer-related gene. SKAP1 expression is significantly increased in colon cancer cells. High SKAP1 levels are independently predictive of poor survival in patients with colon cancer. Notably, SKAP1 expression in colon cancer cells exerted a significant tumor-promoting effect in vivo rather than in vitro. Screening of tumor-infiltrating immune cells revealed the involvement of neutrophils in SKAP1-induced colon tumor promotion. Enhanced formation of neutrophil extracellular traps (NETs) is found to be a key downstream event that contributed to the pro-tumor role of SKAP1. In colon cancer cells, SKAP1 increased the expression of C-X-C motif chemokine ligand 8 (CXCL8) via nuclear factor of activated T cells c1 (NFATc1). The blockade of CXCL8 or NFATc1 largely attenuated neutrophil infiltration, NET formation, and tumor promotion induced by SKAP1. Furthermore, inhibiting SKAP1-induced NET significantly enhanced the antitumor efficiency of adoptive natural killer cell therapy in colon tumor models. In conclusion, SKAP1 significantly promotes colon cancer growth via the cancer cell/neutrophil NFATc1/CXCL8/NET axis, suggesting that SKAP1 is a potential target for colon cancer therapy.
Collapse
Affiliation(s)
- Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Jun Liu
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Jilin Lu
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Xiaofei Zhang
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Wei Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Qian Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Jiayi Cai
- Clinical Research UnitRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Mengjun Li
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Yu Gan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Yifan Tang
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Shuangjie Wu
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| |
Collapse
|
4
|
Evtimov VJ, Hammett MV, Pupovac A, Nguyen NYN, Shu R, Van Der Weyden C, Twigger R, Nisbet IT, Trounson AO, Boyd RL, Prince HM. Targeting TAG-72 in cutaneous T cell lymphoma. Heliyon 2024; 10:e36298. [PMID: 39263154 PMCID: PMC11386021 DOI: 10.1016/j.heliyon.2024.e36298] [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: 03/03/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 09/13/2024] Open
Abstract
Purpose Current monoclonal antibody-based treatment approaches for cutaneous T cell lymphoma (CTCL) rely heavily on the ability to identify a tumor specific target that is essentially absent on normal cells. Herein, we propose tumor associated glycoprotein-72 (TAG-72) as one such target. TAG-72 is a mucin-associated, truncated O-glycan that has been identified as a chimeric antigen receptor (CAR)-T cell target in solid tumor indications. To date, TAG-72 targeting has not been considered in the setting of hematological malignancies. Experimental design CD3+ cells from patients with CTCL were analyzed for TAG-72 expression by flow cytometry. Immunohistochemistry was used to assess TAG-72 expression in CTCL patient skin lesions and a TAG-72 ELISA was employed to assess soluble TAG-72 (CA 72-4) in patient plasma. TAG-72 CAR transduction was performed on healthy donor (HD) and CTCL T cells and characterized by flow cytometry. In vitro CAR-T cell function was assessed by flow cytometry and xCELLigence® using patient peripheral blood mononuclear cells and proof-of-concept ovarian cancer cell lines. In vivo CAR-T cell function was assessed in a proof-of-concept, TAG-72+ ovarian cancer xenograft mouse model. Results TAG-72 expression was significantly higher on total CD3+ T cells and CD4+ subsets in CTCL donors across disease stages, compared to that of HDs. TAG-72 was also present in CTCL patient skin lesions, whereas CA 72-4 was detected at low levels in both CTCL patient and HD plasma with no differences between the two groups. In vitro cytotoxicity assays showed that anti-TAG-72 CAR-T cells significantly, and specifically reduced CD3+TAG-72+ expressing CTCL cells, compared to culture with unedited T cells (no CAR). CTCL CAR-T cells had comparable function to HD CAR-T cells in vitro and CAR-T cells derived from CTCL patients eradicated cancer cells in vivo. Conclusion This study shows the first evidence of TAG-72 as a possible target for the treatment of CTCL.
Collapse
Affiliation(s)
- Vera J Evtimov
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Maree V Hammett
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Aleta Pupovac
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Nhu-Y N Nguyen
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Runzhe Shu
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Carrie Van Der Weyden
- Cartherics Pty Ltd, Notting Hill, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Robert Twigger
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Ian T Nisbet
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Alan O Trounson
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Richard L Boyd
- Cartherics Pty Ltd, Notting Hill, Australia
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - H Miles Prince
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
5
|
Di Prete M, Michelerio A, Lora V, Tomasini CF, Cota C. When mycosis fungoides seems not to be within the spectrum of clinical and histopathological differential diagnoses. Dermatol Reports 2024; 16:10008. [PMID: 39295886 PMCID: PMC11406214 DOI: 10.4081/dr.2024.10008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 04/17/2024] [Indexed: 09/21/2024] Open
Abstract
The most prevalent primary cutaneous T-cell lymphoma, mycosis fungoides (MF), is characterized by the development of plaques and nodules after an erythematous patchy phase that is non-specific. An infiltrate of atypical small- to medium-sized cerebriform lymphocytes in the superficial dermis, with variable epidermotropism, is the histopathological hallmark of the disease. In more advanced stages of the illness, large-cell transformation may be seen. Early diagnosis of MF can be very challenging based only on histopathologic or clinical findings, so it is critical to have a clinical-pathological correlation. Many atypical variants of MF that deviate from the classic Alibert-Bazin presentation of the disease have been described over the past 30 years, sometimes with different prognostic and therapeutic implications. Clinically or histopathologically, they can mimic a wide range of benign inflammatory skin disorders. To make a conclusive diagnosis in these cases, it is recommended to take multiple biopsies from various lesions and to carefully correlate the clinical and pathological findings. We have outlined the various facets of the illness in this review, positioning MF as a "great imitator", with an emphasis on the more recently identified variations, differential diagnosis, and its benign mimics.
Collapse
Affiliation(s)
- Monia Di Prete
- Dermatopathology Laboratory, San Gallicano Dermatological Institute IRCCS, Rome
| | - Andrea Michelerio
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia
- Dermatology Unit, Cardinal Massaia Hospital, Asti
| | - Viviana Lora
- Clinical Dermatology Unit, San Gallicano Dermatological Institute IRCCS, Rome
| | - Carlo Francesco Tomasini
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia
- Dermatology Clinic, IRCCS San Matteo Foundation Hospital, Pavia, Italy
| | - Carlo Cota
- Dermatopathology Laboratory, San Gallicano Dermatological Institute IRCCS, Rome
| |
Collapse
|
6
|
Lachance M, Thibeault MM. Clinical characteristics, initial treatment, and prognosis of mycosis fungoides and Sézary syndrome: A retrospective, single-center study at the Centre Hospitalier Universitaire of Quebec. Ann Dermatol Venereol 2023; 150:276-280. [PMID: 37777355 DOI: 10.1016/j.annder.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/03/2023] [Accepted: 06/26/2023] [Indexed: 10/02/2023]
Affiliation(s)
- M Lachance
- Department of Dermatology, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada.
| | - M-M Thibeault
- Department of Dermatology, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
| |
Collapse
|
7
|
Zhong G, Wu D, Chen H, Yan L, Xiang Q, Liu Y, Wang T. Multi-omics Analysis of Prognostic Significance and Immune Infiltration of FASTK Family Members in Kidney Renal Clear Cell Carcinoma. Evol Bioinform Online 2023; 19:11769343231212078. [PMID: 38033663 PMCID: PMC10683404 DOI: 10.1177/11769343231212078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023] Open
Abstract
Objective The Fas-activated serine/threonine kinase (FASTK) family of proteins has been recently found to be able to regulate mitochondrial gene expression post-transcriptionally. Nonetheless, there is a paucity of study about the role of the FASTK family in kidney renal clear cell carcinoma (KIRC). This study was conducted to explore the correlation of FASTK family genes with expression, prognosis, and immune infiltration in KIRC. Methods We collected the data from the UALCAN, GeneMANIA, STRING, CancerSEA, cBioPortal, Kaplan-Meier plotter, GEPIA, TISIDB and TIMER databases to evaluate the genetic alterations, differential expression, prognostic significance, and immune cell infiltration of FASTKs in patients with KIRC. Results In tumor tissues of KIRC, the mRNA expression level of FASTK and TBRG4 was elevated, whereas that of FASTKD1, FASTKD2, and FASTKD5 was lowered compared with normal tissues (P < .05). Patients with KIRC and high FASTK and Transforming growth factor β regulator 4 (TBRG4) expression had worse overall survival (OS) and disease specific survival (DFS), while those with lower expression of FASTKD2/3/5 had worse outcomes. FASTK was positively correlated with DNA damage. FASTKD1 was positively related to differentiation. FASTKD2 was inversely related to proliferation and FASTKD5 was inversely related to invasion and EMT in KIRC cells. FASTK expression in KIRC was inversely linked to the presence of several immune cells including Tgd, macrophages, Tcm, and Mast cells (P < .05). Conclusions Our research provided fresh insight and in-depth analysis to the selection of prognostic biological markers of FASTK family members in KIRC.
Collapse
Affiliation(s)
- Guanghui Zhong
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Dali Wu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Haiping Chen
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Lingfei Yan
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Qi Xiang
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yufeng Liu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Tao Wang
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Stuver R, Geller S. Advances in the treatment of mycoses fungoides and Sézary syndrome: a narrative update in skin-directed therapies and immune-based treatments. Front Immunol 2023; 14:1284045. [PMID: 37868986 PMCID: PMC10585160 DOI: 10.3389/fimmu.2023.1284045] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Mycoses fungoides (MF) and Sézary syndrome (SS) are cutaneous T-cell lymphomas that are often challenging to manage given the absence of reliably curative therapies, at times high symptom burden with significant detriment to quality of life, and need for ongoing treatment for disease and symptom control. Recent developments in skin-directed treatments include optimizing the use of existing topical therapies, the introduction of known dermatological agents and treatment modalities for the specific treatment of MF/SS (such as mechlorethamine gel, calcineurin inhibitor creams, and photodynamic therapy), and novel local and topical agents. For advanced disease, dedicated clinical trials have translated to exciting progress, leading to the approval of brentuximab vedotin (2017) and mogamulizumab (2018) for relapsed MF/SS. Additional studies of other active systemic agents, including various cellular therapies, represent further attempts to add to the therapeutic armamentarium in treating MF/SS. In this review, we highlight these recent advancements, ranging from optimization of skin-directed therapies to the introduction of novel systemic agents. We focus on therapies approved in the preceding five years or under investigation in advanced-phase clinical trials.
Collapse
Affiliation(s)
- Robert Stuver
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Shamir Geller
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
9
|
Gaillard JB, Chapiro E, Daudignon A, Nadal N, Penther D, Chauzeix J, Nguyen-Khac F, Veronese L, Lefebvre C. Cytogenetics in the management of mature T-cell and NK-cell neoplasms: Guidelines from the Groupe Francophone de Cytogénétique Hématologique (GFCH). Curr Res Transl Med 2023; 71:103428. [PMID: 38016421 DOI: 10.1016/j.retram.2023.103428] [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: 07/04/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
Mature T-cell and natural killer (NK)-cell neoplasms (MTNKNs) are a highly heterogeneous group of lymphomas that represent 10-15 % of lymphoid neoplasms and have usually an aggressive behavior. Diagnosis can be challenging due to their overlapping clinical, histological and immunophenotypic features. Genetic data are not a routine component of the diagnostic algorithm for most MTNKNs. Indeed, unlike B-cell lymphomas, the genomic landscape of MTNKNs is not fully understood. Only few characteristic rearrangements can be easily identified with conventional cytogenetic methods and are an integral part of the diagnostic criteria, for instance the t(14;14)/inv(14) or t(X;14) abnormality harbored by 95 % of patients with T-cell prolymphocytic leukemia, or the ALK gene translocation observed in some forms of anaplastic large cell lymphoma. However, advances in molecular and cytogenetic techniques have brought new insights into MTNKN pathogenesis. Several recurrent genetic alterations have been identified, such as chromosomal losses involving tumor suppressor genes (SETD2, CDKN2A, TP53) and gains involving oncogenes (MYC), activating mutations in signaling pathways (JAK-STAT, RAS), and epigenetic dysregulation, that have improved our understanding of these pathologies. This work provides an overview of the cytogenetics knowledge in MTNKNs in the context of the new World Health Organization classification and the International Consensus Classification of hematolymphoid tumors. It describes key genetic alterations and their clinical implications. It also proposes recommendations on cytogenetic methods for MTNKN diagnosis.
Collapse
Affiliation(s)
- Jean-Baptiste Gaillard
- Unité de Génétique Chromosomique, Service de Génétique moléculaire et cytogénomique, CHU Montpellier, Montpellier, France.
| | - Elise Chapiro
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS_1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; Sorbonne Université, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Service d'Hématologie Biologique, F-75013 Paris, France
| | - Agnès Daudignon
- Institut de Génétique Médicale - Hôpital Jeanne de Flandre - CHRU de Lille, France
| | - Nathalie Nadal
- Service de génétique chromosomique et moléculaire, CHU Dijon, Dijon, France
| | - Dominique Penther
- Laboratoire de Génétique Oncologique, Centre Henri Becquerel, Rouen, France
| | - Jasmine Chauzeix
- Service d'Hématologie biologique CHU de Limoges - CRIBL, UMR CNRS 7276/INSERM 1262, Limoges, France
| | - Florence Nguyen-Khac
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS_1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; Sorbonne Université, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Service d'Hématologie Biologique, F-75013 Paris, France
| | - Lauren Veronese
- Service de Cytogénétique Médicale, CHU Estaing, 1 place Lucie et Raymond Aubrac, 63003 Clermont-Ferrand; EA7453 CHELTER, Université Clermont Auvergne, France
| | - Christine Lefebvre
- Unité de Génétique des Hémopathies, Service d'Hématologie Biologique, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
10
|
Zhu L, Yu Q, Li Y, Zhang M, Peng Z, Wang S, Quan Z, Gao D. SKAP1 Is a Novel Biomarker and Therapeutic Target for Gastric Cancer: Evidence from Expression, Functional, and Bioinformatic Analyses. Int J Mol Sci 2023; 24:11870. [PMID: 37511629 PMCID: PMC10380396 DOI: 10.3390/ijms241411870] [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: 05/15/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related death worldwide. Due to the lack of early symptoms, GC is often diagnosed at an advanced stage when treatment options are limited. There is an urgent need to identify biomarkers for early detection, prognosis evaluation, and targeted treatment of GC. Studies have shown that Src kinase-associated phosphoprotein 1 (SKAP1) promotes cell proliferation and invasion and is associated with poor prognosis in colorectal cancer, malignant fibrous histiocytoma, and breast cancer. However, the role and mechanism of SKAP1 in GC are unclear. Here, analyses of multiple databases and experiments revealed that SKAP1 expression was higher in GC than in adjacent normal tissues. The Cancer Genome Atlas data showed that high SKAP1 expression was associated with poor GC prognosis. SKAP1 expression was higher in GC than in normal gastric epithelial cells. SKAP1 silencing reduced the proliferation, migration and invasion of the GC cell lines MKN45 and HGC27. Rescue experiments suggest that SKAP1 may promote GC progression by activating JAK1/PI3K/AKT signaling and regulating GC cell proliferation, invasion, migration, and other functions. Bioinformatics analysis revealed that SKAP1 was associated with immune cell infiltration and checkpoint expression in GC. High SKAP1 expression was associated with poorer immunotherapy outcomes, suggesting its potential as a predictive biomarker of GC immunotherapy efficacy. In summary, SKAP1 is overexpressed in GC, where it promotes cell proliferation, invasion and migration and is associated with poor prognosis and poor immunotherapy outcomes. SKAP1 may represent a biomarker and therapeutic target in GC and regulates cellular functions through JAK1/PI3K/AKT signaling.
Collapse
Affiliation(s)
- Lingqin Zhu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qiongfang Yu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yuanheng Li
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Meng Zhang
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang 330006, China
| | - Zhiwei Peng
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang 330006, China
| | - Song Wang
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ziyi Quan
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang 330006, China
| | - Dian Gao
- Department of Pathogen Biology and Immunology, Medical College of Nanchang University, Nanchang 330006, China
| |
Collapse
|
11
|
Kalliara E, Belfrage E, Gullberg U, Drott K, Ek S. Spatially Guided and Single Cell Tools to Map the Microenvironment in Cutaneous T-Cell Lymphoma. Cancers (Basel) 2023; 15:cancers15082362. [PMID: 37190290 DOI: 10.3390/cancers15082362] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Mycosis fungoides (MF) and Sézary syndrome (SS) are two closely related clinical variants of cutaneous T-cell lymphomas (CTCL). Previously demonstrated large patient-to-patient and intra-patient disease heterogeneity underpins the importance of personalized medicine in CTCL. Advanced stages of CTCL are characterized by dismal prognosis, and the early identification of patients who will progress remains a clinical unmet need. While the exact molecular events underlying disease progression are poorly resolved, the tumor microenvironment (TME) has emerged as an important driver. In particular, the Th1-to-Th2 shift in the immune response is now commonly identified across advanced-stage CTCL patients. Herein, we summarize the role of the TME in CTCL evolution and the latest studies in deciphering inter- and intra-patient heterogeneity. We introduce spatially resolved omics as a promising technology to advance immune-oncology efforts in CTCL. We propose the combined implementation of spatially guided and single-cell omics technologies in paired skin and blood samples. Such an approach will mediate in-depth profiling of phenotypic and molecular changes in reactive immune subpopulations and malignant T cells preceding the Th1-to-Th2 shift and reveal mechanisms underlying disease progression from skin-limited to systemic disease that collectively will lead to the discovery of novel biomarkers to improve patient prognostication and the design of personalized treatment strategies.
Collapse
Affiliation(s)
- Eirini Kalliara
- Department of Immunotechnology, Faculty of Engineering (LTH), University of Lund, 223 63 Lund, Sweden
| | - Emma Belfrage
- Department of Dermatology and Venereology, Skane University Hospital (SUS), 205 02 Lund, Sweden
| | - Urban Gullberg
- Department of Hematology and Transfusion Medicine, Skane University Hospital (SUS), 205 02 Lund, Sweden
| | - Kristina Drott
- Department of Hematology and Transfusion Medicine, Skane University Hospital (SUS), 205 02 Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Faculty of Engineering (LTH), University of Lund, 223 63 Lund, Sweden
| |
Collapse
|
12
|
An update on genetic aberrations in T-cell neoplasms. Pathology 2023; 55:287-301. [PMID: 36801152 DOI: 10.1016/j.pathol.2022.12.350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 01/20/2023]
Abstract
T-cell neoplasms are a highly heterogeneous group of leukaemias and lymphomas that represent 10-15% of all lymphoid neoplasms. Traditionally, our understanding of T-cell leukaemias and lymphomas has lagged behind that of B-cell neoplasms, in part due to their rarity. However, recent advances in our understanding of T-cell differentiation, based on gene expression and mutation profiling and other high throughput methods, have better elucidated the pathogenetic mechanisms of T-cell leukaemias and lymphomas. In this review, we provide an overview of many of the molecular abnormalities that occur in various types of T-cell leukaemia and lymphoma. Much of this knowledge has been used to refine diagnostic criteria that has been included in the fifth edition of the World Health Organization. This knowledge is also being used to improve prognostication and identify novel therapeutic targets, and we expect this progress will continue, eventually resulting in improved outcomes for patients with T-cell leukaemias and lymphomas.
Collapse
|
13
|
Andrades A, Peinado P, Alvarez-Perez JC, Sanjuan-Hidalgo J, García DJ, Arenas AM, Matia-González AM, Medina PP. SWI/SNF complexes in hematological malignancies: biological implications and therapeutic opportunities. Mol Cancer 2023; 22:39. [PMID: 36810086 PMCID: PMC9942420 DOI: 10.1186/s12943-023-01736-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
Hematological malignancies are a highly heterogeneous group of diseases with varied molecular and phenotypical characteristics. SWI/SNF (SWItch/Sucrose Non-Fermentable) chromatin remodeling complexes play significant roles in the regulation of gene expression, being essential for processes such as cell maintenance and differentiation in hematopoietic stem cells. Furthermore, alterations in SWI/SNF complex subunits, especially in ARID1A/1B/2, SMARCA2/4, and BCL7A, are highly recurrent across a wide variety of lymphoid and myeloid malignancies. Most genetic alterations cause a loss of function of the subunit, suggesting a tumor suppressor role. However, SWI/SNF subunits can also be required for tumor maintenance or even play an oncogenic role in certain disease contexts. The recurrent alterations of SWI/SNF subunits highlight not only the biological relevance of SWI/SNF complexes in hematological malignancies but also their clinical potential. In particular, increasing evidence has shown that mutations in SWI/SNF complex subunits confer resistance to several antineoplastic agents routinely used for the treatment of hematological malignancies. Furthermore, mutations in SWI/SNF subunits often create synthetic lethality relationships with other SWI/SNF or non-SWI/SNF proteins that could be exploited therapeutically. In conclusion, SWI/SNF complexes are recurrently altered in hematological malignancies and some SWI/SNF subunits may be essential for tumor maintenance. These alterations, as well as their synthetic lethal relationships with SWI/SNF and non-SWI/SNF proteins, may be pharmacologically exploited for the treatment of diverse hematological cancers.
Collapse
Affiliation(s)
- Alvaro Andrades
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Paola Peinado
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain ,grid.451388.30000 0004 1795 1830Present Address: The Francis Crick Institute, London, UK
| | - Juan Carlos Alvarez-Perez
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Juan Sanjuan-Hidalgo
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
| | - Daniel J. García
- grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.4489.10000000121678994Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Granada, Spain
| | - Alberto M. Arenas
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Ana M. Matia-González
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Pedro P. Medina
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| |
Collapse
|
14
|
Sorger H, Dey S, Vieyra‐Garcia PA, Pölöske D, Teufelberger AR, de Araujo ED, Sedighi A, Graf R, Spiegl B, Lazzeri I, Braun T, Garces de los Fayos Alonso I, Schlederer M, Timelthaler G, Kodajova P, Pirker C, Surbek M, Machtinger M, Graier T, Perchthaler I, Pan Y, Fink‐Puches R, Cerroni L, Ober J, Otte M, Albrecht JD, Tin G, Abdeldayem A, Manaswiyoungkul P, Olaoye OO, Metzelder ML, Orlova A, Berger W, Wobser M, Nicolay JP, André F, Nguyen VA, Neubauer HA, Fleck R, Merkel O, Herling M, Heitzer E, Gunning PT, Kenner L, Moriggl R, Wolf P. Blocking STAT3/5 through direct or upstream kinase targeting in leukemic cutaneous T-cell lymphoma. EMBO Mol Med 2022; 14:e15200. [PMID: 36341492 PMCID: PMC9727928 DOI: 10.15252/emmm.202115200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 09/26/2022] [Accepted: 10/02/2022] [Indexed: 11/09/2022] Open
Abstract
Leukemic cutaneous T-cell lymphomas (L-CTCL) are lymphoproliferative disorders of skin-homing mature T-cells causing severe symptoms and high mortality through chronic inflammation, tissue destruction, and serious infections. Despite numerous genomic sequencing efforts, recurrent driver mutations have not been identified, but chromosomal losses and gains are frequent and dominant. We integrated genomic landscape analyses with innovative pharmacologic interference studies to identify key vulnerable nodes in L-CTCL. We detected copy number gains of loci containing the STAT3/5 oncogenes in 74% (n = 17/23) of L-CTCL, which correlated with the increased clonal T-cell count in the blood. Dual inhibition of STAT3/5 using small-molecule degraders and multi-kinase blockers abolished L-CTCL cell growth in vitro and ex vivo, whereby PAK kinase inhibition was specifically selective for L-CTCL patient cells carrying STAT3/5 gains. Importantly, the PAK inhibitor FRAx597 demonstrated encouraging anti-leukemic activity in vivo by inhibiting tumor growth and disease dissemination in intradermally xenografted mice. We conclude that STAT3/5 and PAK kinase interaction represents a new therapeutic node to be further explored in L-CTCL.
Collapse
Affiliation(s)
- Helena Sorger
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
- Department of Pediatric and Adolescent Surgery, Vienna General HospitalMedical University of ViennaViennaAustria
| | - Saptaswa Dey
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
- Department of PathologyMedical University of ViennaViennaAustria
| | | | - Daniel Pölöske
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | | | - Elvin D de Araujo
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Abootaleb Sedighi
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Ricarda Graf
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Benjamin Spiegl
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Isaac Lazzeri
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Till Braun
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
| | - Ines Garces de los Fayos Alonso
- Department of PathologyMedical University of ViennaViennaAustria
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | | | | | - Petra Kodajova
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Christine Pirker
- Centre for Cancer ResearchMedical University of ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Marta Surbek
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Michael Machtinger
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Thomas Graier
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | | | - Yi Pan
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Regina Fink‐Puches
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Lorenzo Cerroni
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
| | - Jennifer Ober
- Core Facility Flow Cytometry, Center for Medical Research (ZMF)Medical University of GrazGrazAustria
| | - Moritz Otte
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
| | - Jana D Albrecht
- Department of DermatologyUniversity Hospital MannheimMannheimGermany
| | - Gary Tin
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Ayah Abdeldayem
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Pimyupa Manaswiyoungkul
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Olasunkanmi O Olaoye
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
| | - Martin L Metzelder
- Department of Pediatric and Adolescent Surgery, Vienna General HospitalMedical University of ViennaViennaAustria
| | - Anna Orlova
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Walter Berger
- Centre for Cancer ResearchMedical University of ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Marion Wobser
- Department of DermatologyUniversity Hospital WuerzburgWuerzburgGermany
| | - Jan P Nicolay
- Department of DermatologyUniversity Hospital MannheimMannheimGermany
| | - Fiona André
- University Clinic for Dermatology, Venereology and Allergology InnsbruckMedical University of InnsbruckInnsbruckAustria
| | - Van Anh Nguyen
- University Clinic for Dermatology, Venereology and Allergology InnsbruckMedical University of InnsbruckInnsbruckAustria
| | - Heidi A Neubauer
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | | | - Olaf Merkel
- Department of PathologyMedical University of ViennaViennaAustria
| | - Marco Herling
- Department of Medicine ICIO‐ABCD, CECAD and CMMC Cologne UniversityCologneGermany
- Department of Hematology, Cellular Therapy, and HemostaseologyUniversity of LeipzigLeipzigGermany
| | - Ellen Heitzer
- Diagnostic & Research Center for Molecular Bio‐Medicine, Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Patrick T Gunning
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaONCanada
- Centre for Medicinal ChemistryUniversity of Toronto MississaugaMississaugaONCanada
- Janpix, a Centessa CompanyLondonUK
| | - Lukas Kenner
- Department of PathologyMedical University of ViennaViennaAustria
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
- Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for Applied Metabolomics (CDL‐AM), Division of Nuclear MedicineMedical University of ViennaViennaAustria
- CBmed GmbH Center for Biomarker Research in MedicineGrazAustria
| | - Richard Moriggl
- Unit of Functional Cancer Genomics, Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Peter Wolf
- Department of Dermatology and VenereologyMedical University of GrazGrazAustria
- BioTechMed GrazGrazAustria
| |
Collapse
|
15
|
Genetics Abnormalities with Clinical Impact in Primary Cutaneous Lymphomas. Cancers (Basel) 2022; 14:cancers14204972. [PMID: 36291756 PMCID: PMC9599538 DOI: 10.3390/cancers14204972] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary The genetic landscape of cutaneous T-cell lymphomas analyzed by sequencing high throughput techniques shows a heterogeneous somatic mutational profile and genomic copy number variations in the TCR signaling effectors, the NF-κB elements, DNA damage/repair elements, JAK/STAT pathway elements and epigenetic modifiers. A mutational and genomic stratification of these patients provides new opportunities for the development or repurposing of (personalized) therapeutic strategies. The genetic heterogeneity in cutaneous B-cell lymphoma parallels with the specific subtype. Damaging mutations in primary cutaneous diffuse large B-cell lymphoma of the leg type, involving MYD88 gene, or BCL6 and MYC translocations or CDKN2A deletions are useful for diagnostic purposes. The more indolent forms, as the primary cutaneous lymphoma of follicle center cell (somatic mutations in TNFRSF14 and 1p36 deletions) and the cutaneous lymphoproliferative disorder of the marginal zone cells (FAS gene), present with a more restricted pattern of genetic alterations. Abstract Primary cutaneous lymphomas comprise a heterogeneous group of extranodal non-Hodgkin lymphomas (NHL) that arise from skin resident lymphoid cells and are manifested by specific lymphomatous cutaneous lesions with no evidence of extracutaneous disease at the time of diagnosis. They may originate from mature T-lymphocytes (70% of all cases), mature B-lymphocytes (25–30%) or, rarely, NK cells. Cutaneous T-cell lymphomas (CTCL) comprise a heterogeneous group of T-cell malignancies including Mycosis Fungoides (MF) the most frequent subtype, accounting for approximately half of CTCL, and Sézary syndrome (SS), which is an erythrodermic and leukemic subtype characterized by significant blood involvement. The mutational landscape of MF and SS by NGS include recurrent genomic alterations in the TCR signaling effectors (i.e., PLCG1), the NF-κB elements (i.e., CARD11), DNA damage/repair elements (TP53 or ATM), JAK/STAT pathway elements or epigenetic modifiers (DNMT3). Genomic copy number variations appeared to be more prevalent than somatic mutations. Other CTCL subtypes such as primary cutaneous anaplastic large cell lymphoma also harbor genetic alterations of the JAK/STAT pathway in up to 50% of cases. Recently, primary cutaneous aggressive epidermotropic T-cell lymphoma, a rare fatal subtype, was found to contain a specific profile of JAK2 rearrangements. Other aggressive cytotoxic CTCL (primary cutaneous γδ T-cell lymphomas) also show genetic alterations in the JAK/STAT pathway in a large proportion of patients. Thus, CTCL patients have a heterogeneous genetic/transcriptional and epigenetic background, and there is no uniform treatment for these patients. In this scenario, a pathway-based personalized management is required. Cutaneous B-cell lymphoma (CBCL) subtypes present a variable genetic profile. The genetic heterogeneity parallels the multiple types of specialized B-cells and their specific tissue distribution. Particularly, many recurrent hotspot and damaging mutations in primary cutaneous diffuse large B-cell lymphoma of the leg type, involving MYD88 gene, or BCL6 and MYC translocations and BLIMP1 or CDKN2A deletions are useful for diagnostic and prognostic purposes for this aggressive subtype from other indolent CBCL forms.
Collapse
|
16
|
Pu Y, Wei J, Wu Y, Zhao K, Wu Y, Wu S, Yang X, Xing C. THUMPD3-AS1 facilitates cell growth and aggressiveness by the miR-218-5p/SKAP1 axis in colorectal cancer. Cell Biochem Biophys 2022; 80:483-494. [PMID: 35538197 DOI: 10.1007/s12013-022-01074-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/22/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant cancer with a high mortality. Accumulating studies have revealed that mRNAs involved in ceRNA (competing endogenous RNA) network are implicated in the tumorigenesis and development of CRC. Here, we aimed to elucidate the ceRNA network involving Src kinase associated phosphoprotein 1 (SKAP1) in the biological characteristics of CRC. METHODS Expression levels of genes in colon adenocarcinoma (COAD) samples and prognosis of COAD patients were predicted using publicly available online tool. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), clony formation and Transwell assays were conducted to test the biological functions of SKAP1 and THUMPD3 antisense RNA 1 (THUMPD3-AS1) in CRC cells. Western blot was used to measure the protein levels of SKAP1. Gene expression in CRC cells was detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR). The interaction between miR-218-5p and THUMPD3-AS1 (or SKAP1) was verified by RNA pulldown and luciferase reporter assays. RESULTS SKAP1 was upregulated in COAD tissues and CRC cells and it reflected a poor prognosis in patients with COAD. SKAP1 knockdown inhibited CRC (HT-29 and HCT-116) cell proliferation, migration and invasion. Mechanistically, THUMPD3-AS1 acted as a ceRNA to sponge miR-218-5p and subsequently upregulated SKAP1 expression in CRC cells. SKAP1 overexpression reversed the suppressive effect of THUMPD3-AS1 knockdown on proliferation, migration and invision of CRC cells. CONCLUSIONS THUMPD3-AS1 promotes CRC cell growth and aggressiveness by regulating the miR-218-5p/SKAP1 axis.
Collapse
Affiliation(s)
- Yuwei Pu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Jinrong Wei
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yong Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Kui Zhao
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yongyou Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Shu Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xiaodong Yang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| | - Chungen Xing
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
17
|
To V, Evtimov VJ, Jenkin G, Pupovac A, Trounson AO, Boyd RL. CAR-T cell development for Cutaneous T cell Lymphoma: current limitations and potential treatment strategies. Front Immunol 2022; 13:968395. [PMID: 36059451 PMCID: PMC9433932 DOI: 10.3389/fimmu.2022.968395] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T therapy has demonstrated remarkable outcomes for B cell malignancies, however, its application for T cell lymphoma, particularly cutaneous T cell lymphoma (CTCL), has been limited. Barriers to effective CAR-T cell therapy in treating CTCL include T cell aplasia in autologous transplants, CAR-T product contamination with leukemic T cells, CAR-T fratricide (when the target antigen is present on normal T cells), and tumor heterogeneity. To address these critical challenges, innovative CAR engineering by targeting multiple antigens to strike a balance between efficacy and safety of the therapy is necessary. In this review, we discuss the current obstacles to CAR-T cell therapy and highlight potential targets in treating CTCL. Looking forward, we propose strategies to develop more powerful dual CARs that are advancing towards the clinic in CTCL therapy.
Collapse
Affiliation(s)
- Van To
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | | | - Graham Jenkin
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | | | - Alan O. Trounson
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Richard L. Boyd
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- *Correspondence: Richard L. Boyd,
| |
Collapse
|
18
|
Kumar S, Dhamija B, Attrish D, Sawant V, Sengar M, Thorat J, Shet T, Jain H, Purwar R. Genetic alterations and oxidative stress in T cell lymphomas. Pharmacol Ther 2022; 236:108109. [PMID: 35007658 DOI: 10.1016/j.pharmthera.2022.108109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/20/2022]
Abstract
T cell lymphomas encompass a diverse group of Non-Hodgkin lymphomas with a wide spectrum of clinical, immunological and pathological manifestations. In the last two decades there has been a progress in our understanding of the cell of origin, genetic abnormalities and their impact on behaviour in T cell lymphomas. Genetic alterations are one of the critical drivers of the pathogenesis of T cell lymphoma. Disease progression has been correlated with multiple genetic abnormalities where malignant clones arise primarily out of the host immune surveillance arsenal. There are many cellular processes involved in disease development, and some of them are T cell signaling, differentiation, epigenetic modifications, and immune regulation. Modulation of these crucial pathways via genetic mutations and chromosomal abnormalities possessing either point or copy number mutations helps tumor cells to develop a niche favourable for their growth via metabolic alterations. Several metabolic pathways especially regulation of redox homeostasis is critical in pathogenesis of lymphoma. Disruption of redox potential and induction of oxidative stress renders malignant cells vulnerable to mitochondrial damage and triggers apoptotic pathways causing cell death. Targeting genetic abnormalities and oxidative stress along with current treatment regime have the potential for improved therapeutics and presents new combination approaches towards selective treatment of T cell lymphomas.
Collapse
Affiliation(s)
- Sushant Kumar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Bhavuk Dhamija
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Diksha Attrish
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Vinanti Sawant
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Manju Sengar
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Jayashree Thorat
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Tanuja Shet
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Hasmukh Jain
- Medical Oncology, Tata memorial Hospital, Mumbai, Maharashtra 400012, India
| | - Rahul Purwar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India.
| |
Collapse
|
19
|
Gaydosik AM, Stonesifer CJ, Khaleel AE, Geskin LJ, Fuschiotti P. Single-Cell RNA Sequencing Unveils the Clonal and Transcriptional Landscape of Cutaneous T-Cell Lymphomas. Clin Cancer Res 2022; 28:2610-2622. [PMID: 35421230 PMCID: PMC9197926 DOI: 10.1158/1078-0432.ccr-21-4437] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/24/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE Clonal malignant T lymphocytes constitute only a fraction of T cells in mycosis fungoides skin tumors and in the leukemic blood of Sézary syndrome, the classic types of cutaneous T-cell lymphomas. However, lack of markers specific for malignant lymphocytes prevents distinguishing them from benign T cells, thus delaying diagnosis and the development of targeted treatments. Here we applied single-cell methods to assess the transcriptional profiles of both malignant T-cell clones and reactive T lymphocytes directly in mycosis fungoides/Sézary syndrome patient samples. EXPERIMENTAL DESIGN Single-cell RNA sequencing was used to profile the T-cell immune repertoire simultaneously with gene expression in CD3+ lymphocytes from mycosis fungoides and healthy skin biopsies as well as from Sézary syndrome and control blood samples. Transcriptional data were validated in additional advanced-stage mycosis fungoides/Sézary syndrome skin and blood samples by immunofluorescence microscopy. RESULTS Several nonoverlapping clonotypes are expanded in the skin and blood of individual advanced-stage mycosis fungoides/Sézary syndrome patient samples, including a dominant malignant clone as well as additional minor malignant and reactive clones. While we detected upregulation of patient-specific as well as mycosis fungoides- and Sézary syndrome-specific oncogenic pathways within individual malignant clones, we also detected upregulation of several common pathways that included genes associated with cancer cell metabolism, cell-cycle regulation, de novo nucleotide biosynthesis, and invasion. CONCLUSIONS Our analysis unveils new insights into mycosis fungoides/Sézary syndrome pathogenesis by providing an unprecedented report of the transcriptional profile of malignant T-cell clones in the skin and blood of individual patients and offers novel prospective targets for personalized therapy.
Collapse
Affiliation(s)
- Alyxzandria M. Gaydosik
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA 15261, USA
| | | | | | | | - Patrizia Fuschiotti
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA 15261, USA,Correspondence to: Patrizia Fuschiotti, Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, S709 BST, 200 Lothrop Street, Pittsburgh PA 15261, USA. Tel.: +1-412-648-9385;
| |
Collapse
|
20
|
Karagianni F, Piperi C, Casar B, de la Fuente-Vivas D, García-Gómez R, Lampadaki K, Pappa V, Papadavid E. Combination of Resminostat with Ruxolitinib Exerts Antitumor Effects in the Chick Embryo Chorioallantoic Membrane Model for Cutaneous T Cell Lymphoma. Cancers (Basel) 2022; 14:cancers14041070. [PMID: 35205818 PMCID: PMC8870185 DOI: 10.3390/cancers14041070] [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: 01/25/2022] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary The combination of Resminostat (HDACi) and Ruxolitinib (JAKi) exerted cytotoxic effects and inhibited proliferation of CTCL cell lines (MyLa, SeAx) in vitro. The aim of the present study was to validate their antitumor effects in vivo using the chick embryo chorioallantoic membrane (CAM) model, which allows quick and efficient monitoring of tumor growth, migration, invasion, and metastatic potential. The drug combination exhibited a significant inhibition of primary tumor size, and inhibited intravasation and extravasation of tumor cells to the liver and lung. It also exerted an inhibitory effect in the migration and invasion of tumor cells and significantly reduced key signaling pathway activation. Our data demonstrate that the CAM assay could be employed as a preclinical in vivo model in CTCL for pharmacological testing, and that the combination of Resminostat and Ruxolitinib exerts significant antitumor effects in CTCL progression that need to be further evaluated in a clinical setting. Abstract The combination of Resminostat (HDACi) and Ruxolitinib (JAKi) exerted cytotoxic effects and inhibited proliferation of CTCL cell lines (MyLa, SeAx) in previously published work. A xenograft tumor formation was produced by implanting the MyLa or SeAx cells on top of the chick embryo chorioallantoic membrane (CAM). The CAM assay protocol was developed to monitor the metastatic properties of CTCL cells and the effects of Resminostat and/or Ruxolitinib in vivo. In the spontaneous CAM assays, Resminostat and Ruxolitinib treatment inhibited the cell proliferation (p < 0.001) of MyLa and SeAx, and induced cell apoptosis (p < 0.005, p < 0.001, respectively). Although monotherapies reduced the size of primary tumors in the metastasis CAM assay, the drug combination exhibited a significant inhibition of primary tumor size (p < 0.0001). Furthermore, the combined treatment inhibited the intravasation of MyLa (p < 0.005) and SeAx cells (p < 0.0001) in the organs, as well as their extravasation to the liver (p < 0.0001) and lung (p < 0.0001). The drug combination also exerted a stronger inhibitory effect in migration (p < 0.0001) rather in invasion (p < 0.005) of both MyLa and SeAx cells. It further reduced p-p38, p-ERK, p-AKT, and p-STAT in MyLa cells, while it decreased p-ERK and p-STAT in SeAx cells in CAM tumors. Our data demonstrated that the CAM assay could be employed as a preclinical in vivo model in CTCL for pharmacological testing. In agreement with previous in vitro data, the combination of Resminostat and Ruxolitinib was shown to exert antitumor effects in CTCL in vivo.
Collapse
Affiliation(s)
- Fani Karagianni
- National Center of Rare Diseases-Cutaneous Lymphoma—Member of EuroBloodNet, Second Department of Dermatology and Venereal Diseases, Attikon University General Hospital, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (F.K.); (K.L.)
| | - Christina Piperi
- Department of Biological Chemistry, Medical School of Athens, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Cantabria, 39011 Santander, Spain; (D.d.l.F.-V.); (R.G.-G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (B.C.); (E.P.)
| | - Dalia de la Fuente-Vivas
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Cantabria, 39011 Santander, Spain; (D.d.l.F.-V.); (R.G.-G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rocío García-Gómez
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Cantabria, 39011 Santander, Spain; (D.d.l.F.-V.); (R.G.-G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Kyriaki Lampadaki
- National Center of Rare Diseases-Cutaneous Lymphoma—Member of EuroBloodNet, Second Department of Dermatology and Venereal Diseases, Attikon University General Hospital, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (F.K.); (K.L.)
| | - Vasiliki Pappa
- 2nd Department of Internal Medicine—Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School of Athens, University General Hospital Attikon, 124 62 Athens, Greece;
| | - Evangelia Papadavid
- National Center of Rare Diseases-Cutaneous Lymphoma—Member of EuroBloodNet, Second Department of Dermatology and Venereal Diseases, Attikon University General Hospital, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (F.K.); (K.L.)
- Correspondence: (B.C.); (E.P.)
| |
Collapse
|
21
|
Gill RPK, Gantchev J, Martínez Villarreal A, Ramchatesingh B, Netchiporouk E, Akilov OE, Ødum N, Gniadecki R, Koralov SB, Litvinov IV. Understanding Cell Lines, Patient-Derived Xenograft and Genetically Engineered Mouse Models Used to Study Cutaneous T-Cell Lymphoma. Cells 2022; 11:cells11040593. [PMID: 35203244 PMCID: PMC8870189 DOI: 10.3390/cells11040593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Cutaneous T cell lymphoma (CTCL) is a spectrum of lymphoproliferative disorders caused by the infiltration of malignant T cells into the skin. The most common variants of CTCL include mycosis fungoides (MF), Sézary syndrome (SS) and CD30+ Lymphoproliferative disorders (CD30+ LPDs). CD30+ LPDs include primary cutaneous anaplastic large cell lymphoma (pcALCL), lymphomatoid papulosis (LyP) and borderline CD30+ LPD. The frequency of MF, SS and CD30+ LPDs is ~40–50%, <5% and ~10–25%, respectively. Despite recent advances, CTCL remains challenging to diagnose. The mechanism of CTCL carcinogenesis still remains to be fully elucidated. Hence, experiments in patient-derived cell lines and xenografts/genetically engineered mouse models (GEMMs) are critical to advance our understanding of disease pathogenesis. To enable this, understanding the intricacies and limitations of each individual model system is highly important. Presently, 11 immortalized patient-derived cell lines and different xenograft/GEMMs are being used to study the pathogenesis of CTCL and evaluate the therapeutic efficacy of various treatment modalities prior to clinical trials. Gene expression studies, and the karyotyping analyses of cell lines demonstrated that the molecular profile of SeAx, Sez4, SZ4, H9 and Hut78 is consistent with SS origin; MyLa and HH resemble the molecular profile of advanced MF, while Mac2A and PB2B represent CD30+ LPDs. Molecular analysis of the other two frequently used Human T-Cell Lymphotropic Virus-1 (HTLV-1)+ cell lines, MJ and Hut102, were found to have characteristics of Adult T-cell Leukemia/Lymphoma (ATLL). Studies in mouse models demonstrated that xenograft tumors could be grown using MyLa, HH, H9, Hut78, PB2B and SZ4 cells in NSG (NOD Scid gamma mouse) mice, while several additional experimental GEMMs were established to study the pathogenesis, effect of drugs and inflammatory cytokines in CTCL. The current review summarizes cell lines and xenograft/GEMMs used to study and understand the etiology and heterogeneity of CTCL.
Collapse
Affiliation(s)
- Raman Preet Kaur Gill
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Jennifer Gantchev
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Amelia Martínez Villarreal
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Brandon Ramchatesingh
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Elena Netchiporouk
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
| | - Oleg E. Akilov
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Niels Ødum
- Division of Dermatology, University of Alberta, Edmonton, AB T6G 2B7, Canada;
| | - Robert Gniadecki
- Skin Immunology Research Center, University of Copenhagen, DK-2200 Copenhagen, Denmark;
| | - Sergei B. Koralov
- Department of Pathology, New York University, New York, NY 10016, USA;
| | - Ivan V. Litvinov
- Division of Dermatology, McGill University, Montreal, QC H4A 3J1, Canada; (R.P.K.G.); (J.G.); (A.M.V.); (B.R.); (E.N.)
- Correspondence: ; Tel.: +514-934-1934 (ext. 76140); Fax: +514-843-1570
| |
Collapse
|
22
|
Liu F, Gao Y, Xu B, Xiong S, Yi S, Sun J, Chen Z, Liu X, Li Y, Lin Y, Wen Y, Qin Y, Yang S, Li H, Tejasvi T, Tsoi L, Tu P, Ren X, Wang Y. PEG10 amplification at 7q21.3 potentiates large-cell transformation in cutaneous T-cell lymphoma. Blood 2022; 139:554-571. [PMID: 34582557 PMCID: PMC8893588 DOI: 10.1182/blood.2021012091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/07/2021] [Indexed: 01/29/2023] Open
Abstract
Mycosis fungoides (MF), the most common form of cutaneous T-cell lymphoma, undergo large-cell transformation (LCT) in the late stage, manifesting aggressive behavior, resistance to treatments, and poor prognosis, but the mechanisms involved remain unclear. To identify the molecular driver of LCT, we collected tumor samples from 133 MF patients and performed whole-transcriptome sequencing on 49 advanced-stage MF patients, followed by integrated copy number inference and genomic hybridization. Tumors with LCT showed unique transcriptional programs and enriched expressions of genes at chr7q. Paternally expressed gene 10 (PEG10), an imprinted gene at 7q21.3, was ectopically expressed in malignant T cells from LCT, driven by 7q21.3 amplification. Mechanistically, aberrant PEG10 expression increased cell size, promoted cell proliferation, and conferred treatment resistance by a PEG10/KLF2/NF-κB axis in in vitro and in vivo models. Pharmacologically targeting PEG10 reversed the phenotypes of proliferation and treatment resistance in LCT. Our findings reveal new molecular mechanisms underlying LCT and suggest that PEG10 inhibition may serve as a promising therapeutic approach in late-stage aggressive T-cell lymphoma.
Collapse
MESH Headings
- Animals
- Apoptosis Regulatory Proteins/genetics
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- DNA-Binding Proteins/genetics
- Female
- Gene Amplification
- Gene Expression Regulation, Neoplastic
- Genomic Imprinting
- Humans
- Lymphoma, T-Cell, Cutaneous/genetics
- Lymphoma, T-Cell, Cutaneous/pathology
- Mice, Inbred NOD
- Mice, SCID
- Mycosis Fungoides/genetics
- Mycosis Fungoides/pathology
- RNA-Binding Proteins/genetics
- Skin Neoplasms/genetics
- Skin Neoplasms/pathology
- Mice
Collapse
Affiliation(s)
- Fengjie Liu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yumei Gao
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Bufang Xu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Shan Xiong
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Shengguo Yi
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Jingru Sun
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Zhuojing Chen
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xiangjun Liu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yingyi Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yuchieh Lin
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yujie Wen
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yao Qin
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Shuxia Yang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Hang Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Trilokraj Tejasvi
- Department of Dermatology, University of Michigan, Ann Arbor, MI; and
| | - Lam Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI; and
| | - Ping Tu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100034, China
| | - Yang Wang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| |
Collapse
|
23
|
Licht P, Mailänder V. Transcriptional Heterogeneity and the Microbiome of Cutaneous T-Cell Lymphoma. Cells 2022; 11:cells11030328. [PMID: 35159138 PMCID: PMC8834405 DOI: 10.3390/cells11030328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cutaneous T-Cell Lymphomas (CTCL) presents with substantial clinical variability and transcriptional heterogeneity. In the recent years, several studies paved the way to elucidate aetiology and pathogenesis of CTCL using sequencing methods. Several T-cell subtypes were suggested as the source of disease thereby explaining clinical and transcriptional heterogeneity of CTCL entities. Several differentially expressed pathways could explain disease progression. However, exogenous triggers in the skin microenvironment also seem to affect CTCL status. Especially Staphylococcus aureus was shown to contribute to disease progression. Only little is known about the complex microbiome patterns involved in CTCL and how microbial shifts might impact this malignancy. Nevertheless, first hints indicate that the microbiome might at least in part explain transcriptional heterogeneity and that microbial approaches could serve in diagnosis and prognosis. Shaping the microbiome could be a treatment option to maintain stable disease. Here, we review current knowledge of transcriptional heterogeneity of and microbial influences on CTCL. We discuss potential benefits of microbial applications and microbial directed therapies to aid patients with CTCL burden.
Collapse
Affiliation(s)
- Philipp Licht
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Volker Mailänder
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Correspondence:
| |
Collapse
|
24
|
Fujii K. Pathogenesis of cutaneous T cell lymphoma: Involvement of Staphylococcus aureus. J Dermatol 2021; 49:202-209. [PMID: 34927279 DOI: 10.1111/1346-8138.16288] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/30/2022]
Abstract
Mycosis fungoides (MF) and Sézary syndrome (SS) are representative cutaneous lymphomas. In their early stage, a small number of tumor cells and a large number of non-malignant cells form a Th1-dominant tumor microenvironment. Increase in malignant T cells is accompanied by a decrease in CD8-positive T cells, with a shift toward a Th2-dominant milieu in advanced-stage lesions. The etiologies of MF/SS are diverse, and the underlying pathogenetic mechanisms are yet to be elucidated. Advanced MF/SS is known to be highly sensitive to Staphylococcus aureus, and the majority of deaths are caused by severe infections. The susceptibility to infection is associated with barrier dysfunction and immunosuppression, which are the main symptoms of MF. In recent years, skin-colonizing S. aureus has been identified to not only cause severe infections but also play an important role in the pathogenesis of MF/SS. Staphylococcal superantigens activate the proliferation of tumor cells and induce CD25 upregulation, FOXP3 expression, IL-17 expression, and miR-155 expression. Alpha-toxin eliminates non-neoplastic CD4-positive cells and CD8-positive cells and plays a major role in tumor cell selection. Lipoprotein may also be associated with the induction of Th2-dominant milieu. Antibiotic therapy for S. aureus eradication has been reported to cause considerable clinical improvement in the majority of individuals with advanced cutaneous T-cell lymphoma. Therefore, S. aureus may be a novel target for the treatment of advanced-stage MF/SS in the future.
Collapse
Affiliation(s)
- Kazuyasu Fujii
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
25
|
Liu Z, Wu X, Hwang ST, Liu J. The Role of Tumor Microenvironment in Mycosis Fungoides and Sézary Syndrome. Ann Dermatol 2021; 33:487-496. [PMID: 34858000 PMCID: PMC8577908 DOI: 10.5021/ad.2021.33.6.487] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 11/25/2022] Open
Abstract
Mycosis fungoides (MF) and Sézary syndrome (SS) are the most common subtypes of cutaneous T-cell lymphomas (CTCLs). Most cases of MF display an indolent course during its early stage. However, in some patients, it can progress to the tumor stage with potential systematic involvement and a poor prognosis. SS is defined as an erythrodermic CTCL with leukemic involvements. The pathogenesis of MF and SS is still not fully understood, but recent data have found that the development of MF and SS is related to genetic alterations and possibly to environmental influences. In CTCL, many components interacting with tumor cells, such as tumor-associated macrophages, fibroblasts, dendritic cells, mast cells, and myeloid-derived suppressor cells, as well as with chemokines, cytokines and other key players, establish the tumor microenvironment (TME). In turn, the TME regulates tumor cell migration and proliferation directly and indirectly and may play a critical role in the progression of MF and SS. The TME of MF and SS appear to show features of a Th2 phenotype, thus dampening tumor-related immune responses. Recently, several studies have been published on the immunological characteristics of MF and SS, but a full understanding of the CTCL-related TME remains to be determined. This review focuses on the role of the TME in MF and SS, aiming to further demonstrate the pathogenesis of the disease and to provide new ideas for potential treatments targeted at the microenvironment components of the tumor.
Collapse
Affiliation(s)
- Zhaorui Liu
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Xuesong Wu
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA, United States
| | - Sam T Hwang
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA, United States
| | - Jie Liu
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| |
Collapse
|
26
|
Cuesta-Mateos C, Terrón F, Herling M. CCR7 in Blood Cancers - Review of Its Pathophysiological Roles and the Potential as a Therapeutic Target. Front Oncol 2021; 11:736758. [PMID: 34778050 PMCID: PMC8589249 DOI: 10.3389/fonc.2021.736758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
According to the classical paradigm, CCR7 is a homing chemokine receptor that grants normal lymphocytes access to secondary lymphoid tissues such as lymph nodes or spleen. As such, in most lymphoproliferative disorders, CCR7 expression correlates with nodal or spleen involvement. Nonetheless, recent evidence suggests that CCR7 is more than a facilitator of lymphatic spread of tumor cells. Here, we review published data to catalogue CCR7 expression across blood cancers and appraise which classical and novel roles are attributed to this receptor in the pathogenesis of specific hematologic neoplasms. We outline why novel therapeutic strategies targeting CCR7 might provide clinical benefits to patients with CCR7-positive hematopoietic tumors.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto la Princesa (IIS-IP), Madrid, Spain.,Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Fernando Terrón
- Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Marco Herling
- Clinic of Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
27
|
Leupin N, Zinzani PL, Morschhauser F, Dalle S, Maerevoet M, Michot JM, Ribrag V, Offner F, Beylot-Barry M, Moins-Teisserenc H, Zwaenepoel K, de Winne K, Battistella M, Hultberg A, Gandini D, Moshir M, Jacobs J, Delahaye T, Khan A, Zabrocki P, Silence K, van Rompaey L, Borg C, Motta G, Melle F, Calleri A, Pauwels P, de Haard H, Pileri S, Bagot M. Cusatuzumab for treatment of CD70-positive relapsed or refractory cutaneous T-cell lymphoma. Cancer 2021; 128:1004-1014. [PMID: 34726773 DOI: 10.1002/cncr.34005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/15/2021] [Accepted: 10/01/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND The clinical benefit of cusatuzumab, a CD70-directed monoclonal antibody with enhanced effector functions, was investigated in patients with relapsed/refractory (R/R) cutaneous T-cell lymphoma (CTCL). METHODS In this cohort expansion of the ARGX-110-1201 study, 27 patients with R/R CTCL received cusatuzumab at 1 (n = 11) or 5 mg/kg (n = 16) once every 3 weeks to investigate its safety, dose, and exploratory efficacy. The pharmacokinetics, immunogenicity, CD70 expression, and CD70/CD27 biology were also assessed. RESULTS The most common adverse events included infusion-related reactions, pyrexia, and asthenia. Eighteen serious adverse events (grade 1-3) were reported in 11 patients; 1 of these (vasculitis) was considered drug-related. For 8 of the 11 patients receiving 1 mg/kg, anti-drug antibodies (ADAs) affected the minimal concentration, and this resulted in undetectable cusatuzumab concentrations at the end of treatment and, in some cases, a loss of response. This effect was greatly reduced in the patients receiving 5 mg/kg. The overall response rate was 23%; this included 1 complete response and 5 partial responses (PRs) in 26 of the 27 evaluable patients. In addition, 9 patients achieved stable disease. The mean duration on cusatuzumab was 5.2 months, and the median duration was 2.5 months. Patients with Sézary syndrome (SS) achieved a 60% PR rate with a dosage of 5 mg/kg and a 33% PR rate with a dosage of 1 mg/kg; this resulted in an overall response rate of 50% for patients with SS at both doses. CONCLUSIONS Cusatuzumab was well tolerated, and antitumor activity was observed at both 1 and 5 mg/kg in highly pretreated patients with R/R CTCL. The observed dose-dependent effect on exposure supports the use of 5 mg/kg for future development.
Collapse
Affiliation(s)
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Istituto di Ematologia "Seràgnoli," Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale Università degli Studi, Bologna, Italy
| | - Franck Morschhauser
- Groupe de Recherche sur les Formes Injectables et les Technologies Associées, Université de Lille, CHU Lille, EA 7365, Lille, France
| | - Stéphane Dalle
- Department of Dermatology, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Marie Maerevoet
- Service Hématologie, Institut Jules Bordet, Brussels, Belgium
| | | | | | | | - Marie Beylot-Barry
- Inserm U1053, Department of Dermatology, Centre Hospitalier, Bordeaux, France
| | | | - Karen Zwaenepoel
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Koen de Winne
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | | | | | | | | | | | | | | | | | | | | | - Christophe Borg
- Inserm U645, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Giovanna Motta
- Division of Hematopathology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Federica Melle
- Division of Hematopathology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Angelica Calleri
- Division of Hematopathology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Patrick Pauwels
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | | | - Stefano Pileri
- Division of Hematopathology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Martine Bagot
- Inserm U976, Hôpital Saint Louis, Université de Paris, Paris, France
| |
Collapse
|
28
|
Herrera A, Cheng A, Mimitou EP, Seffens A, George D, Bar-Natan M, Heguy A, Ruggles KV, Scher JU, Hymes K, Latkowski JA, Ødum N, Kadin ME, Ouyang Z, Geskin LJ, Smibert P, Buus TB, Koralov SB. Multimodal single-cell analysis of cutaneous T-cell lymphoma reveals distinct subclonal tissue-dependent signatures. Blood 2021; 138:1456-1464. [PMID: 34232982 PMCID: PMC8532199 DOI: 10.1182/blood.2020009346] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/03/2021] [Indexed: 11/20/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a heterogeneous group of mature T-cell neoplasms characterized by the accumulation of clonal malignant CD4+ T cells in the skin. The most common variant of CTCL, mycosis fungoides (MF ), is confined to the skin in early stages but can be accompanied by extracutaneous dissemination of malignant T cells to the blood and lymph nodes in advanced stages of disease. Sézary syndrome (SS), a leukemic form of disease, is characterized by significant blood involvement. Little is known about the transcriptional and genomic relationship between skin- and blood-residing malignant T cells in CTCL. To identify and interrogate malignant clones in matched skin and blood from patients with leukemic MF and SS, we combine T-cell receptor clonotyping with quantification of gene expression and cell surface markers at the single cell level. Our data reveal clonal evolution at a transcriptional and genetic level within the malignant populations of individual patients. We highlight highly consistent transcriptional signatures delineating skin- and blood-derived malignant T cells. Analysis of these 2 populations suggests that environmental cues, along with genetic aberrations, contribute to transcriptional profiles of malignant T cells. Our findings indicate that the skin microenvironment in CTCL promotes a transcriptional response supporting rapid malignant expansion, as opposed to the quiescent state observed in the blood, potentially influencing efficacy of therapies. These results provide insight into tissue-specific characteristics of cancerous cells and underscore the need to address the patients' individual malignant profiles at the time of therapy to eliminate all subclones.
Collapse
Affiliation(s)
- Alberto Herrera
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Anthony Cheng
- Department of Genetic and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA
| | - Eleni P Mimitou
- Technology Innovation Laboratory, New York Genome Center, New York, NY
| | - Angelina Seffens
- Department of Pathology, New York University School of Medicine, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Dean George
- Department of Dermatology, Boston University and Roger Williams Medical Center, Brown University, Providence, RI
| | - Michal Bar-Natan
- Department of Pathology, New York University School of Medicine, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adriana Heguy
- Department of Pathology, New York University School of Medicine, New York, NY
- Genome Technology Center, New York University School of Medicine, New York, NY
| | | | - Jose U Scher
- Division of Rheumatology, Department of Medicine
| | | | - Jo-Ann Latkowski
- Department of Dermatology, New York University School of Medicine, New York, NY
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Marshall E Kadin
- Department of Dermatology, Boston University and Roger Williams Medical Center, Brown University, Providence, RI
| | - Zhengqing Ouyang
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA
| | - Larisa J Geskin
- Department of Dermatology, Columbia University, New York, NY
| | - Peter Smibert
- Technology Innovation Laboratory, New York Genome Center, New York, NY
| | - Terkild B Buus
- Department of Pathology, New York University School of Medicine, New York, NY
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, NY
| |
Collapse
|
29
|
Magraner-Pardo L, Gobelli D, de la Fuente MA, Pons T, Simarro M. Systematic Analysis of FASTK Gene Family Alterations in Cancer. Int J Mol Sci 2021; 22:11337. [PMID: 34768773 PMCID: PMC8583194 DOI: 10.3390/ijms222111337] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/28/2022] Open
Abstract
The FASTK family of proteins have been recently reported to play a key role in the post-transcriptional regulation of mitochondrial gene expression, including mRNA stability and translation. Accumulated studies have provided evidence that the expression of some FASTK genes is altered in certain types of cancer, in agreement with the central role of mitochondria in cancer development. Here, we obtained a pan-cancer overview of the genomic and transcriptomic alterations of FASTK genes. FASTK, FASTKD1, FASTKD3 and FASTKD5 showed the highest rates of genetic alterations. FASTK and FASTKD3 alterations consisted mainly of amplifications that were seen in more than 8% of ovarian and lung cancers, respectively. FASTKD1 and FASTKD5 were the most frequently mutated FASTK genes, and the mutations were identified in 5-7% of uterine cancers, as well as in 4% of melanomas. Our results also showed that the mRNA levels of all FASTK members were strongly upregulated in esophageal, stomach, liver and lung cancers. Finally, the protein-protein interaction network for FASTK proteins uncovers the interaction of FASTK, FASTKD2, FASTKD4 and FASTKD5 with cancer signaling pathways. These results serve as a starting point for future research into the potential of the FASTK family members as diagnostic and therapeutic targets for certain types of cancer.
Collapse
Affiliation(s)
- Lorena Magraner-Pardo
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain;
| | - Dino Gobelli
- Department of Cell Biology, Histology and Pharmacology, University of Valladolid, 47005 Valladolid, Spain; (D.G.); (M.A.d.l.F.)
- Unit of Excellence, Institute of Biology and Molecular Genetics, University of Valladolid and CSIC, 47003 Valladolid, Spain
| | - Miguel A. de la Fuente
- Department of Cell Biology, Histology and Pharmacology, University of Valladolid, 47005 Valladolid, Spain; (D.G.); (M.A.d.l.F.)
- Unit of Excellence, Institute of Biology and Molecular Genetics, University of Valladolid and CSIC, 47003 Valladolid, Spain
| | - Tirso Pons
- Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Spanish National Research Council, 28049 Madrid, Spain;
| | - María Simarro
- Unit of Excellence, Institute of Biology and Molecular Genetics, University of Valladolid and CSIC, 47003 Valladolid, Spain
- Department of Department of Nursing, University of Valladolid, 47005 Valladolid, Spain
| |
Collapse
|
30
|
Genetic and epigenetic insights into cutaneous T-cell lymphoma. Blood 2021; 139:15-33. [PMID: 34570882 DOI: 10.1182/blood.2019004256] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/30/2021] [Indexed: 11/20/2022] Open
Abstract
Primary cutaneous T-cell lymphomas (CTCL) constitute a heterogeneous group of non-Hodgkin T-cell lymphomas that present in the skin. In recent years significant progress has been made in the understanding of the pathogenesis of CTCL. Progress in CTCL classifications combined with technical advances, in particular next generation sequencing (NGS), enabled a more detailed analysis of the genetic and epigenetic landscape and transcriptional changes in clearly defined diagnostic entities. These studies not only demonstrated extensive heterogeneity between different CTCL subtypes but also identified recurrent alterations that are highly characteristic for diagnostic subgroups of CTCL. The identified alterations in particular involve epigenetic remodelling, cell cycle regulation, and the constitutive activation of targetable, oncogenic pathways. In this respect, aberrant JAK-STAT signaling is a recurrent theme, however not universal for all CTCL and with seemingly different underlaying causes in different entities. A number of the mutated genes identified are potentially actionable targets for the development of novel therapeutic strategies. Moreover, these studies have produced an enormous amount of information that will be critically important for the further development of improved diagnostic and prognostic biomarkers that can assist in the clinical management of CTCL patients. In the present review the main findings of these studies in relation to their functional impact on the malignant transformation process are discussed for different subtypes of CTCL.
Collapse
|
31
|
Xiao MZX, Hennessey D, Iyer A, O'Keefe S, Zhang F, Sivanand A, Gniadecki R. Transcriptomic Changes During Stage Progression of Mycosis Fungoides. Br J Dermatol 2021; 186:520-531. [PMID: 34528236 DOI: 10.1111/bjd.20760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mycosis fungoides (MF) is the most common cutaneous T cell lymphoma, which in the early patch/plaque stages runs an indolent course. However, ~25% of MF patients develop skin tumors, a hallmark of progression to the advanced stage and is associated with high mortality. The mechanisms involved in stage progression are poorly elucidated. METHODS We performed whole-transcriptome and whole-exome sequencing of malignant MF cells from skin biopsies obtained by laser-capture microdissection. We compared three types of MF lesions: early-stage plaques (ESP, n=12) as well as plaques and tumors from patients in late-stage disease (late-stage plaques [LSP], n=10, and tumors [TMR], n=15). Gene Ontology (GO) and KEGG analysis were used to determine pathway changes specific for different lesions which were linked to the recurrent somatic mutations overrepresented in MF tumors. RESULTS The key upregulated pathways during stage progression were those related to cell proliferation and survival (MEK/ERK, Akt-mTOR), Th2/Th9 signaling (IL4, STAT3, STAT5, STAT6), meiomitosis (CT45A1, CT45A3, STAG3, GTSF1, REC8) and DNA repair (PARP1, MYCN, OGG1). Principal coordinate clustering of the transcriptome revealed extensive gene expression differences between early (ESP) and advanced-stage lesions (LSP and TMR). LSP and TMR showed remarkable similarities at the level of the transcriptome, which we interpreted as evidence of cell percolation between lesions via hematogenous self-seeding. CONCLUSION Stage progression in MF is associated with Th2/Th9 polarization of malignant cells, activation of proliferation, survival, as well as increased genomic instability. Global transcriptomic changes in multiple lesions may be caused by hematogenous cell percolation between discrete skin lesions.
Collapse
Affiliation(s)
- M Z X Xiao
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - D Hennessey
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - A Iyer
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - S O'Keefe
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - F Zhang
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - A Sivanand
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - R Gniadecki
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
32
|
Abstract
Primary cutaneous T cell lymphomas (CTCLs) are a heterogeneous group of lymphomas that present in the skin with no evidence of extracutaneous disease at the time of diagnosis. CTCL subtypes demonstrate a variety of clinical, histological, and molecular features, and can follow an indolent or a very aggressive course. The underlying pathogenetic mechanisms are not yet entirely understood. The pathophysiology of CTCL is complex and a single initiating factor has not yet been identified. Diagnosis is based on clinicopathological correlation and requires an interdisciplinary team. Treatment decision is made based on short-term and long-term goals. Therapy options comprise skin-directed therapies, such as topical steroids or phototherapy, and systemic therapies, such as monoclonal antibodies or chemotherapy. So far, the only curative treatment approach is allogeneic haematopoietic stem cell transplantation. Novel therapies, such as chimeric antigen receptor T cells, monoclonal antibodies or small molecules, are being investigated in clinical trials. Patients with CTCL have reduced quality of life and a lack of effective treatment options. Further research is needed to better identify the underlying mechanisms of CTCL development and course as well as to better tailor treatment strategies to individual patients.
Collapse
|
33
|
Lin M, Kowolik CM, Xie J, Yadav S, Overman LE, Horne DA. Potent Anticancer Effects of Epidithiodiketopiperazine NT1721 in Cutaneous T Cell Lymphoma. Cancers (Basel) 2021; 13:cancers13133367. [PMID: 34282785 PMCID: PMC8268131 DOI: 10.3390/cancers13133367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/24/2021] [Accepted: 07/01/2021] [Indexed: 01/19/2023] Open
Abstract
Simple Summary Cutaneous T cell lymphomas (CTCLs) are a group of blood cancers that cannot be cured with current chemotherapeutical or biological drugs. Patients with advanced disease are severely immunocompromised due to the unchecked expansion of malignant T cells and have low survival rates of less than four years. Hence, new treatment options for CTCLs are urgently needed. In this study the anti-CTCL activity of a new compound, NT1721, was determined in vitro and in two CTCL mouse models. We found that NT1721 increased apoptosis (programmed cell death) in the malignant T cells and reduced tumor growth better than two drugs that are currently clinically used for CTCL treatment (i.e., gemcitabine, romidepsin). These results suggest that NT1721 may represent a potent new agent for the treatment of advanced CTCL. Abstract Cutaneous T cell lymphomas (CTCLs) are a heterogeneous group of debilitating, incurable malignancies. Mycosis fungoides (MF) and Sézary syndrome (SS) are the most common subtypes, accounting for ~65% of CTCL cases. Patients with advanced disease have a poor prognosis and low median survival rates of four years. CTCLs develop from malignant skin-homing CD4+ T cells that spread to lymph nodes, blood, bone marrow and viscera in advanced stages. Current treatments options for refractory or advanced CTCL, including chemotherapeutic and biological approaches, rarely lead to durable responses. The exact molecular mechanisms of CTCL pathology remain unclear despite numerous genomic and gene expression profile studies. However, apoptosis resistance is thought to play a major role in the accumulation of malignant T cells. Here we show that NT1721, a synthetic epidithiodiketopiperazine based on a natural product, reduced cell viability at nanomolar concentrations in CTCL cell lines, while largely sparing normal CD4+ cells. Treatment of CTCL cells with NT1721 reduced proliferation and potently induced apoptosis. NT1721 mediated the downregulation of GLI1 transcription factor, which was associated with decreased STAT3 activation and the reduced expression of downstream antiapoptotic proteins (BCL2 and BCL-xL). Importantly, NT1721, which is orally available, reduced tumor growth in two CTCL mouse models significantly better than two clinically used drugs (romidepsin, gemcitabine). Moreover, a combination of NT1721 with gemcitabine reduced the tumor growth significantly better than the single drugs. Taken together, these results suggest that NT1721 may be a promising new agent for the treatment of CTCLs.
Collapse
Affiliation(s)
- Min Lin
- Department of Molecular Medicine, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA; (M.L.); (J.X.); (S.Y.)
| | - Claudia M. Kowolik
- Department of Molecular Medicine, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA; (M.L.); (J.X.); (S.Y.)
- Correspondence: (C.M.K.); (D.A.H.)
| | - Jun Xie
- Department of Molecular Medicine, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA; (M.L.); (J.X.); (S.Y.)
| | - Sushma Yadav
- Department of Molecular Medicine, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA; (M.L.); (J.X.); (S.Y.)
- Department of Translational Research and Cellular Therapeutics, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Larry E. Overman
- Department of Chemistry, University of California, Irvine, CA 92697-2025, USA;
| | - David A. Horne
- Department of Molecular Medicine, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA; (M.L.); (J.X.); (S.Y.)
- Correspondence: (C.M.K.); (D.A.H.)
| |
Collapse
|
34
|
Motamedi M, Xiao MZX, Iyer A, Gniadecki R. Patterns of Gene Expression in Cutaneous T-Cell Lymphoma: Systematic Review of Transcriptomic Studies in Mycosis Fungoides. Cells 2021; 10:cells10061409. [PMID: 34204115 PMCID: PMC8229125 DOI: 10.3390/cells10061409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023] Open
Abstract
Mycosis fungoides (MF) is the most prevalent type of skin lymphoma. In its early stages, it has a favorable prognosis. However, in its late stages, it is associated with an increased risk of mortality. This systematic review aimed to identify the transcriptomic changes involved in MF pathogenesis and progression. A literature search was conducted using the database PubMed, followed by the extraction of 2245 genes which were further filtered to 150 recurrent genes that appeared in two or more publications. Categorization of these genes identified activated pathways involved in pathways such as cell cycle and proliferation, chromosomal instability, and DNA repair. We identified 15 genes implicated in MF progression, which were involved in cell proliferation, immune checkpoints, resistance to apoptosis, and immune response. In highlighting the discrepancies in the way MF transcriptomic data is obtained, further research can focus on not only unifying their approach but also focus on the 150 pertinent genes identified in this review.
Collapse
Affiliation(s)
- Melika Motamedi
- Division of Dermatology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.M.); (M.Z.X.X.); (A.I.)
| | - Maggie Z. X. Xiao
- Division of Dermatology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.M.); (M.Z.X.X.); (A.I.)
| | - Aishwarya Iyer
- Division of Dermatology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.M.); (M.Z.X.X.); (A.I.)
| | - Robert Gniadecki
- Division of Dermatology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.M.); (M.Z.X.X.); (A.I.)
- 8-112 Clinical Sciences Building, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Correspondence: ; Tel.: +1-(780)-407-1555
| |
Collapse
|
35
|
Gu X, Wang Y, Zhang C, Liu Y. GFI-1 overexpression promotes cell proliferation and apoptosis resistance in mycosis fungoides by repressing Bax and P21. Oncol Lett 2021; 22:521. [PMID: 34025788 PMCID: PMC8130034 DOI: 10.3892/ol.2021.12782] [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: 11/21/2020] [Accepted: 03/10/2021] [Indexed: 11/17/2022] Open
Abstract
Mycosis fungoides (MF) is the most common type of cutaneous T-cell lymphoma. The majority of patients with advanced stage MF are resistant to conventional chemotherapy and thus have a poor prognosis. The transcriptional repressor growth factor independence-1 (GFI-1) serves an important role in the development of T-cells. The results of the present study demonstrated that the expression of GFI-1 at different clinical stages of MF was significantly higher compared with benign inflammatory dermatoses, and there was a significant association with disease progression. Gene knockdown of GFI-1 results in the inhibition of Hut-78 cell proliferation and clone formation in vitro, cell cycle arrest and spontaneous apoptosis, upregulation of cell cycle-related P21, as well as the apoptosis-related proteins Bax and Caspase-3, and downregulation of CDK2. Using luciferase assays, and mutational analysis, it was demonstrated that GFI-1 directly regulated the transcription of P21. The results of the present study highlighted a potential molecular therapeutic approach for the treatment of advanced MF.
Collapse
Affiliation(s)
- Xiaoguang Gu
- Department of Dermatology and Venerology, Aviation General Hospital, Beijing 100012, P.R. China.,Institute of Translational Medicine, Chinese Academy of Sciences, Beijing 100012, P.R. China
| | - Yimeng Wang
- Department of Dermatology and Venerology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Chunlei Zhang
- Department of Dermatology and Venerology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Yongsheng Liu
- Department of Dermatology and Venerology, Aviation General Hospital, Beijing 100012, P.R. China.,Institute of Translational Medicine, Chinese Academy of Sciences, Beijing 100012, P.R. China
| |
Collapse
|
36
|
Karagianni F, Piperi C, Mpakou V, Spathis A, Foukas PG, Dalamaga M, Pappa V, Papadavid E. Ruxolitinib with resminostat exert synergistic antitumor effects in Cutaneous T-cell Lymphoma. PLoS One 2021; 16:e0248298. [PMID: 33705488 PMCID: PMC7951910 DOI: 10.1371/journal.pone.0248298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/23/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The combination of JAK/STAT and HDAC inhibitors exerted beneficial effects in haematological malignancies, presenting promising therapeutic CTCL targets. We aim to investigate the efficacy of JAK1/2i ruxolitinib in combination with HDACi resminostat in CTCL in vitro. MATERIAL & METHODS Non-toxic concentrations of ruxolitinib and/or resminostat were administered to MyLa (MF) and SeAx (SS) cells for 24h. Cytotoxicity, cell proliferation and apoptosis were estimated through MTT, BrdU/7AAD and Annexin V/PI assay. Multi-pathway analysis was performed to investigate the effect of JAK1/2i and/or HDACi on JAK/STAT, Akt/mTOR and MAPK signalling pathways. RESULTS Both drugs and their combination were cytotoxic in MyLa (p<0.05) and in SeAx cell line (p<0.001), inhibited proliferation of MyLa (p<0.001) and SeAx (p<0.001) at 24h, compared to untreated cells. Moreover, combined drug treatment induced apoptosis after 24h (p<0.001) in MyLa, and SeAx (p<0.001). The combination of drugs had a strong synergistic effect with a CI<1. Importantly, the drugs' combination inhibited phosphorylation of STAT3 (p<0.001), Akt (p<0.05), ERK1/2 (p<0.001) and JNK (p<0.001) in MyLa, while it reduced activation of Akt (p<0.05) and JNK (p<0.001) in SeAx. CONCLUSION The JAKi/HDACi combination exhibited substantial anti-tumor effects in CTCL cell lines, and may represent a promising novel therapeutic modality for CTCL patients.
Collapse
Affiliation(s)
- Fani Karagianni
- 2nd Department of Dermatology and Venereal Diseases, NKUA, Athens, Greece
- * E-mail:
| | | | - Vassiliki Mpakou
- Second Department of Internal Medicine and Research Institute, Attikon University General Hospital, NKUA, Athens, Greece
| | - Aris Spathis
- Second Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis G. Foukas
- Second Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Dalamaga
- 2nd Department of Dermatology and Venereal Diseases, NKUA, Athens, Greece
- Department of Biological Chemistry, NKUA, Athens, Greece
| | - Vasiliki Pappa
- Second Department of Internal Medicine and Research Institute, Attikon University General Hospital, NKUA, Athens, Greece
| | | |
Collapse
|
37
|
Durgin JS, Weiner DM, Wysocka M, Rook AH. The immunopathogenesis and immunotherapy of cutaneous T cell lymphoma: Pathways and targets for immune restoration and tumor eradication. J Am Acad Dermatol 2021; 84:587-595. [PMID: 33352267 PMCID: PMC7897252 DOI: 10.1016/j.jaad.2020.12.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 11/27/2022]
Abstract
Cutaneous T cell lymphomas (CTCLs) are malignancies of skin-trafficking T cells. Patients with advanced CTCL manifest immune dysfunction that predisposes to infection and suppresses the antitumor immune response. Therapies that stimulate immunity have produced superior progression-free survival compared with conventional chemotherapy, reinforcing the importance of addressing the immune deficient state in the care of patients with CTCL. Recent research has better defined the pathogenesis of these immune deficits, explaining the mechanisms of disease progression and revealing potential therapeutic targets. The features of the malignant cell in mycosis fungoides and Sézary syndrome are now significantly better understood, including the T helper 2 cell phenotype, regulatory T cell cytokine production, immune checkpoint molecule expression, chemokine receptors, and interactions with the microenvironment. The updated model of CTCL immunopathogenesis provides understanding into clinical progression and therapeutic response.
Collapse
Affiliation(s)
- Joseph S Durgin
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David M Weiner
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria Wysocka
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alain H Rook
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
38
|
Durgin JS, Jariwala NN, Wysocka M, Zhang KK, Maity A, Benoit B, Plastaras JP, Lewis DJ, Rosenthal JM, Teague JE, Berg S, Del Guzzo C, Kim EJ, Vittorio C, Haun PL, Samimi SS, Villasenor-Park J, Inverso J, Clark RA, Rook AH. Low-Dose Total Skin Electron Beam Therapy as Part of a Multimodality Regimen for Treatment of Sézary Syndrome: Clinical, Immunologic, and Molecular Analysis. JAMA Dermatol 2021; 157:90-95. [PMID: 33112366 PMCID: PMC7593882 DOI: 10.1001/jamadermatol.2020.3958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/02/2020] [Indexed: 11/14/2022]
Abstract
Importance Sézary syndrome (SS) is an advanced form of cutaneous T-cell lymphoma with few long-term remissions observed. Objective To profile 3 patients with SS who have experienced long-term remission following the addition of low-dose total skin electron beam therapy (TSEBT) to systemic regimens of extracorporeal photopheresis, bexarotene, and interferon-γ. Design, Setting, and Participants This is a retrospective case series with additional investigations of patient-donated samples to assess therapeutic response. The study was conducted at the University of Pennsylvania Cutaneous Lymphoma Clinic and follows 3 patients with stage IVA1 CD4+ SS who presented to the clinic between November 1, 2009, and November 1, 2017, and who had a history of SS that was refractory to multimodality systemic therapy prior to receiving low-dose TSEBT. Interventions Patients were treated in a multimodality fashion with combined extracorporeal photopheresis, bexarotene, interferon-γ, and low-dose TSEBT. Main Outcomes and Measures To characterize treatment responses in these patients, the extent of skin disease was measured with the modified severity weighted assessment tool. Blood disease was measured with flow cytometric assessments of Sézary cell count, CD4:CD8 ratio, and high throughput sequencing of the T-cell receptors. To assess for restoration of immune function, we measured markers of immune exhaustion, including PD-1 (programmed cell death 1), TIGIT (T-cell immunoreceptor with immunoglobulin and ITIM domains), CTLA4 (cytotoxic T-lymphocyte-associated protein 4), TOX (thymocyte selection-associated high mobility group box protein), and Foxp3 (forkhead box P3) on circulating CD4 and CD8 T cells, along with production capacity of interferon-γ by lymphocytes following activation stimuli. Results Following administration of low-dose TSEBT and maintenance of the other therapies, remissions ranged from 24 to 30 months, with complete responses in 2 patients ongoing. Markers of immune exhaustion including PD-1, TIGIT, CTLA4, TOX, and Foxp3 were significantly reduced from baseline following TSEBT, along with enhanced production capacity of interferon-γ by lymphocytes following activation stimuli. High throughput sequencing demonstrated near-complete eradication of the circulating clone among 2 of 3 patients with stable levels in 1. Conclusions and Relevance We describe 3 patients who achieved long-term clinical and molecular remissions following low-dose TSEBT as part of a multimodality regimen for treatment of SS. As long-term remissions in SS are uncommon, this approach demonstrates promise, and clinical trials should be considered.
Collapse
Affiliation(s)
- Joseph S. Durgin
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Neha N. Jariwala
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Maria Wysocka
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Kevin K. Zhang
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Amit Maity
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Bernice Benoit
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - John P. Plastaras
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J. Lewis
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jaclyn M. Rosenthal
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jessica E. Teague
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Berg
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Christina Del Guzzo
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ellen J. Kim
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Carmela Vittorio
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Paul L. Haun
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Sara S. Samimi
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jennifer Villasenor-Park
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Joanne Inverso
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Rachael A. Clark
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alain H. Rook
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
39
|
Blackmon AL, Pinter-Brown L. Spotlight on Mogamulizumab-Kpkc for Use in Adults with Relapsed or Refractory Mycosis Fungoides or Sézary Syndrome: Efficacy, Safety, and Patient Selection. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3747-3754. [PMID: 32982179 PMCID: PMC7502391 DOI: 10.2147/dddt.s185896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/16/2020] [Indexed: 11/23/2022]
Abstract
Advanced cutaneous T cell lymphomas (CTCL) including mycosis fungoides (MF) and Sézary syndrome (SS) are often difficult to manage once they become resistant to initial systemic treatment. Current systemic treatments usually provide a limited duration of disease control, leaving this an area in desperate need of new treatment options for better long-term control. These conditions often affect the older population where transplantation may not be a feasible option. Recent studies evaluated a novel CCR4 humanized monoclonal antibody, mogamulizumab, in relapsed/refractory MF and SS, which show a meaningful progression free survival (PFS) benefit. In August 2018, mogamulizumab was approved by the FDA for the treatment of patients with relapsed/refractory MF/SS who have failed at least one treatment. Approval was based on the Phase III MAVORIC study comparing mogamulizumab to vorinostat, an FDA approved drug for this indication, in 372 patients. In this trial, mogamulizumab was found to have a superior PFS with a median of 7.7 months compared to 3.1 months in the vorinostat arm, with a hazard ratio of 0.53, p<0.001. Mogamulizumab was well tolerated with the most common AE being infusion-related reactions (32%), drug rash (20%), diarrhea (23%), and fatigue (22%). We reviewed the literature leading to the development and approval of mogamulizumab and suggest which patients may benefit the most from this treatment.
Collapse
Affiliation(s)
- Amanda L Blackmon
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Lauren Pinter-Brown
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
40
|
Bobrowicz M, Fassnacht C, Ignatova D, Chang YT, Dimitriou F, Guenova E. Pathogenesis and Therapy of Primary Cutaneous T-Cell Lymphoma: Collegium Internationale Allergologicum (CIA) Update 2020. Int Arch Allergy Immunol 2020; 181:733-745. [PMID: 32690848 DOI: 10.1159/000509281] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 11/19/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a heterogeneous disease group of unknown etiology with a complex immunological background. As CTCL arises from T cells that have a vital role in the antitumor response, their therapy is largely aimed at reversing the immunological mechanisms leading to or manifesting during this malignancy. Early disease stages can be controlled with skin-directed therapy in most CTCL cases. Still, advanced CTCL has a dismal prognosis and warrants systemic therapy. Despite considerable progress in understanding the pathophysiology of the disease and the numerous systemic treatment options available, long-term remission rates with conventional treatments alone are still low. Allogeneic hematopoietic stem cell transplantation is currently the only curative option for advanced CTCL, including mycosis fungoides and Sézary syndrome. The aims of this review is to summarize the recent findings on the immunology of this heterogeneous disease and to present the advances in its clinical management.
Collapse
Affiliation(s)
| | - Christina Fassnacht
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Desislava Ignatova
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Yun-Tsan Chang
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Department of Dermatology, Lausanne University Hospital (CHUV), Lausanne, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland, .,Faculty of Medicine, University of Zurich, Zurich, Switzerland, .,Department of Dermatology, Lausanne University Hospital (CHUV), Lausanne, Switzerland, .,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,
| |
Collapse
|
41
|
Mirza AS, Horna P, Teer JK, Song J, Akabari R, Hussaini M, Sokol L. New Insights Into the Complex Mutational Landscape of Sézary Syndrome. Front Oncol 2020; 10:514. [PMID: 32373524 PMCID: PMC7186303 DOI: 10.3389/fonc.2020.00514] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 03/23/2020] [Indexed: 12/28/2022] Open
Abstract
Sézary syndrome (SS) is a genetically and clinically distinct entity among cutaneous T-cell lymphomas (CTCL). SS is characterized by more aggressive disease compared to the most common indolent type of CTCL, mycosis fungoides. However, there are limited available genomic data regarding SS. To characterize and expand current mappings of the genomic landscape of CTCL, whole exome sequencing (WES) was performed on peripheral blood samples from seven patients with SS. We detected 21,784 variants, of which 21,140 were novel and 644 were previously described. Filtering revealed 551 nonsynonymous variants among 525 mutated genes−25 recurrent mutations and 1 recurrent variant. Several recurrently mutated genes crucial to pathogenesis pathways, including Janus kinase (JAK)/signal transducers and activators of transcription (STAT), peroxisome proliferator-activated receptors (PPAR), PI3K-serine/threonine protein kinases (AKT), and fibroblast growth factor receptors (FGFR), were identified. Furthermore, genetic mutations spanned both known and novel genes, supporting the idea of a long-tail distribution of mutations in lymphoma. Acknowledging these genetic variants and their affected pathways may inspire future targeted therapies. WES of a limited number of SS patients revealed both novel findings and corroborated complexities of the “long-tail” distribution of previously reported mutations.
Collapse
Affiliation(s)
- Abu-Sayeef Mirza
- Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Pedro Horna
- Division of Hematopathology, Mayo Clinic, Rochester, MN, United States
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Jinming Song
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Ratilal Akabari
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Mohammad Hussaini
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Lubomir Sokol
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
42
|
Larocca C, Kupper TS, LeBoeuf NR. Mogamulizumab Forecast: Clearer Patients, with a Slight Chance of Immune Mayhem. Clin Cancer Res 2019; 25:7272-7274. [PMID: 31615932 PMCID: PMC7147994 DOI: 10.1158/1078-0432.ccr-19-2742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/26/2019] [Accepted: 10/10/2019] [Indexed: 11/16/2022]
Abstract
Mogamulizumab, approved by the FDA for relapsed or refractory mycosis fungoides and Sézary syndrome, improves progression-free survival compared with vorinostat in the largest trial to date in cutaneous T-cell lymphoma, with particular efficacy in leukemic disease, but carries a risk of immune-mediated toxicities with concomitant depletion of regulatory T cells.See related article by Kasamon et al., p. 7275.
Collapse
Affiliation(s)
- Cecilia Larocca
- Department of Dermatology, Brigham and Women's Hospital, and Dana Farber Cancer Institute, Boston, Massachusetts
| | - Thomas S Kupper
- Department of Dermatology, Brigham and Women's Hospital, and Dana Farber Cancer Institute, Boston, Massachusetts.
| | - Nicole R LeBoeuf
- Department of Dermatology, Brigham and Women's Hospital, and Dana Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
43
|
Hu T, Krejsgaard T, Nastasi C, Buus TB, Nansen A, Hald A, Spee P, Nielsen PR, Blümel E, Gluud M, Willerslev-Olsen A, Woetmann A, Bzorek M, Eriksen JO, Ødum N, Rahbek Gjerdrum LM. Expression of the Voltage-Gated Potassium Channel Kv1.3 in Lesional Skin from Patients with Cutaneous T-Cell Lymphoma and Benign Dermatitis. Dermatology 2019; 236:123-132. [PMID: 31536992 DOI: 10.1159/000502137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/11/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The voltage-gated potassium channel Kv1.3 (KCNA3) is expressed by effector memory T cells (TEM) and plays an important role in their activation and proliferation. Mycosis fungoides (MF), the most common subtype of cutaneous T-cell lymphoma (CTCL), was recently proposed to be a malignancy of skin-resident TEM. However, the expression of Kv1.3 in CTCL has not been investigated. OBJECTIVES This study aims to examine the expression of Kv1.3 in situ and in vitro in CTCL. METHODS The expression of Kv1.3 was examined by immunohistochemistry in skin lesions from 38 patients with MF, 4 patients with Sézary syndrome (SS), and 27 patients with benign dermatosis. In 4 malignant T-cell lines of CTCL (Myla2059, PB2B, SeAx, and Mac2a) and a non-malignant T-cell line (MyLa1850), the expression of Kv1.3 was determined by flow cytometry. The proliferation of those cell lines treated with various concentrations of Kv1.3 inhibitor ShK was measured by 3H-thymdine incorporation. RESULTS Half of the MF patients (19/38) displayed partial Kv1.3 expression including 1 patient with moderate Kv1.3 positivity, while the other half (19/38) exhibited Kv1.3 negativity. An almost identical distribution was observed in patients with benign conditions, that is, 44.4% (12/27) were partially positive for Kv1.3 including 1 patient with moderate Kv1.3 positivity, while 55.6% (15/27) were Kv1.3 negative. In contrast, 3 in 4 SS patients displayed partial Kv1.3 positivity including 2 patients with weak staining and 1 with moderate staining, while 1 in 4 SS patients was Kv1.3 negative. In addition, all malignant T-cell lines, and a non-malignant T-cell line, displayed low Kv1.3 surface expression with a similar pattern. Whereas 2 cell lines (PB2B and Mac2a) were sensitive to Kv1.3 blockade, the other 2 (Myla2059 and SeAx) were completely resistant. CONCLUSIONS We provide the first evidence of a heterogeneous Kv1.3 expression in situ in CTCL lesions.
Collapse
Affiliation(s)
- Tengpeng Hu
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Thorbjørn Krejsgaard
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Claudia Nastasi
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Terkild Brink Buus
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anneline Nansen
- Department of in vivo Pharmacology, Zealand Pharma A/S, Glostrup, Denmark
| | - Andreas Hald
- Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Center, University of Copenhagen, Copenhagen, Denmark
| | | | - Pia Rude Nielsen
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Edda Blümel
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Gluud
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Willerslev-Olsen
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anders Woetmann
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Bzorek
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Jens O Eriksen
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
44
|
Moosic KB, Paila U, Olson KC, Dziewulska K, Wang TT, Xing JC, Ratan A, Feith DJ, Loughran TP, Olson TL. Genomics of LGL leukemia and select other rare leukemia/lymphomas. Best Pract Res Clin Haematol 2019; 32:196-206. [PMID: 31585620 PMCID: PMC6779335 DOI: 10.1016/j.beha.2019.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/04/2019] [Indexed: 01/04/2023]
Abstract
Genomic analysis of cancer offers the hope of identifying new treatments or aiding in the selection of existing treatments. Rare leukemias pose additional challenges in this regard as samples may be hard to acquire and when found the underlying pathway may not be attractive to drug development since so few individuals are affected. In this case, it can be useful to identify common mutational overlap among subsets of rare leukemias to increase the number of individuals that may benefit from a targeted therapy. This chapter examines the current mutational landscape of large granular lymphocyte (LGL) leukemia with a focus on STAT3 mutations, the most common mutation in LGL leukemia to date. We examined the linkage between these mutations and autoimmune symptoms and disorders, in cases of obvious and suspected LGL leukemia. We then summarized and compared mutations in a set of other rare leukemias that also have JAK/STAT signaling pathway activation brought about by genomic changes. These include T-cell acute lymphoblastic leukemia (T-ALL), T-cell prolymphocytic leukemia (T-PLL), cutaneous T-cell lymphoma (CTCL), select peripheral T-cell lymphoma (PTCL), and adult T-cell leukemia/lymphoma (ATLL). Though STAT3 activation is common in these leukemias, the way in which it is achieved, such as the activating cytokine pathway and/or the co-mutational background, is quite diverse.
Collapse
Affiliation(s)
- Katharine B Moosic
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Pathology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA.
| | - Umadevi Paila
- Center for Public Health Genomics, MSB-6111A, West Complex, 1335 Lee Street, Charlottesville, VA, 22908, USA.
| | - Kristine C Olson
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA.
| | - Karolina Dziewulska
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Pathology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA.
| | - T Tiffany Wang
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Microbiology, Immunology, and Cancer Biology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA.
| | - Jeffrey C Xing
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| | - Aakrosh Ratan
- Center for Public Health Genomics, MSB-6131F, West Complex, 1300 JPA, Charlottesville, VA, 22908, USA.
| | - David J Feith
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA.
| | - Thomas P Loughran
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA.
| | - Thomas L Olson
- University of Virginia Cancer Center, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA; Department of Medicine, Division of Hematology/Oncology, 345 Crispell Dr, PO Box 801378, Charlottesville, VA, 22908, USA.
| |
Collapse
|
45
|
Borcherding N, Voigt AP, Liu V, Link BK, Zhang W, Jabbari A. Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression. Clin Cancer Res 2019; 25:2996-3005. [PMID: 30718356 PMCID: PMC6659117 DOI: 10.1158/1078-0432.ccr-18-3309] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/07/2018] [Accepted: 01/25/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Cutaneous T-cell lymphomas (CTCL), encompassing a spectrum of T-cell lymphoproliferative disorders involving the skin, have collectively increased in incidence over the last 40 years. Sézary syndrome is an aggressive form of CTCL characterized by significant presence of malignant cells in both the blood and skin. The guarded prognosis for Sézary syndrome reflects a lack of reliably effective therapy, due, in part, to an incomplete understanding of disease pathogenesis. EXPERIMENTAL DESIGN Using single-cell sequencing of RNA and the machine-learning reverse graph embedding approach in the Monocle package, we defined a model featuring distinct transcriptomic states within Sézary syndrome. Gene expression used to differentiate the unique transcriptional states were further used to develop a boosted tree classification for early versus late CTCL disease. RESULTS Our analysis showed the involvement of FOXP3 + malignant T cells during clonal evolution, transitioning from FOXP3 + T cells to GATA3 + or IKZF2 + (HELIOS) tumor cells. Transcriptomic diversities in a clonal tumor can be used to predict disease stage, and we were able to characterize a gene signature that predicts disease stage with close to 80% accuracy. FOXP3 was found to be the most important factor to predict early disease in CTCL, along with another 19 genes used to predict CTCL stage. CONCLUSIONS This work offers insight into the heterogeneity of Sézary syndrome, providing better understanding of the transcriptomic diversities within a clonal tumor. This transcriptional heterogeneity can predict tumor stage and thereby offer guidance for therapy.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Andrew P Voigt
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Vincent Liu
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Department of Dermatology, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Brian K Link
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Weizhou Zhang
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Interdisciplinary Program in Immunology, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Ali Jabbari
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, Iowa.
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Department of Dermatology, University of Iowa, College of Medicine, Iowa City, Iowa
- Interdisciplinary Program in Immunology, University of Iowa, College of Medicine, Iowa City, Iowa
| |
Collapse
|
46
|
Gug G, Huang Q, Chiticariu E, Solovan C, Baudis M. DNA copy number imbalances in primary cutaneous lymphomas. J Eur Acad Dermatol Venereol 2019; 33:1062-1075. [PMID: 30659659 DOI: 10.1111/jdv.15442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cutaneous lymphomas (CL) represent a clinically defined group of extranodal non-Hodgkin lymphomas harbouring heterogeneous and incompletely delineated molecular aberrations. Over the past decades, molecular studies have identified several chromosomal aberrations, but the interpretation of individual genomic studies can be challenging. OBJECTIVE With a comprehensive meta-analysis, we aim to delineate genomic alterations for different types of CL and propose a more accurate classification in line with their various pathogenicity. METHODS We searched PubMed and ISI Web of Knowledge for publications from 1996 to 2016 reporting the investigation of CL for genome-wide copy number alterations, by means of comparative genomic hybridization techniques and whole-genome sequencing and whole-exome sequencing. We then extracted and remapped the available copy number variation (CNV) data from these publications with the same pipeline and performed clustering and visualisation to aggregate samples of similar CNV profiles. RESULTS For 449 samples from 22 publications, CNV data were accessible for sample based meta-analysis. Our findings illustrate structural and numerical chromosomal imbalance patterns. Most frequent CNAs were linked to oncogenes or tumour suppressor genes with important roles in the course of the disease. CONCLUSION Summary profiles for genomic imbalances, generated from case-specific data, identified complex genomic imbalances, which could discriminate between different subtypes of CL and promise a more accurate classification. The collected data presented in this study are publicly available through the 'Progenetix' online repository.
Collapse
Affiliation(s)
- G Gug
- University of Medicine and Pharmacy "Victor Babeș", Timișoara, România
| | - Q Huang
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - E Chiticariu
- University of Medicine and Pharmacy "Victor Babeș", Timișoara, România
| | - C Solovan
- University of Medicine and Pharmacy "Victor Babeș", Timișoara, România.,Emergency City Hospital, University Clinic of Dermatology and Venereology, Timișoara, România
| | - M Baudis
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Swiss Institute of Bioinformatics, Zurich, Switzerland
| |
Collapse
|
47
|
Bastidas Torres AN, Cats D, Mei H, Szuhai K, Willemze R, Vermeer MH, Tensen CP. Genomic analysis reveals recurrent deletion of JAK-STAT signaling inhibitors HNRNPK and SOCS1 in mycosis fungoides. Genes Chromosomes Cancer 2018; 57:653-664. [PMID: 30144205 PMCID: PMC6282857 DOI: 10.1002/gcc.22679] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 01/31/2023] Open
Abstract
Mycosis fungoides (MF) is the most common cutaneous T-cell lymphoma (CTCL). Causative genetic alterations in MF are unknown. The low recurrence of pathogenic small-scale mutations (ie, nucleotide substitutions, indels) in the disease, calls for the study of additional aspects of MF genetics. Here, we investigated structural genomic alterations in tumor-stage MF by integrating whole-genome sequencing and RNA-sequencing. Multiple genes with roles in cell physiology (n = 113) and metabolism (n = 92) were found to be impacted by genomic rearrangements, including 47 genes currently implicated in cancer. Fusion transcripts involving genes of interest such as DOT1L, KDM6A, LIFR, TP53, and TP63 were also observed. Additionally, we identified recurrent deletions of genes involved in cell cycle control, chromatin regulation, the JAK-STAT pathway, and the PI-3-K pathway. Remarkably, many of these deletions result from genomic rearrangements. Deletion of tumor suppressors HNRNPK and SOCS1 were the most frequent genetic alterations in MF after deletion of CDKN2A. Notably, SOCS1 deletion could be detected in early-stage MF. In agreement with the observed genomic alterations, transcriptome analysis revealed up-regulation of the cell cycle, JAK-STAT, PI-3-K and developmental pathways. Our results position inactivation of HNRNPK and SOCS1 as potential driver events in MF development.
Collapse
Affiliation(s)
| | - Davy Cats
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Karoly Szuhai
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rein Willemze
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarten H Vermeer
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis P Tensen
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
48
|
Fanoni D, Corti L, Alberti-Violetti S, Tensen CP, Venegoni L, Vermeer M, Willemze R, Berti E. Array-based CGH of primary cutaneous CD8+ aggressive EPIDERMO-tropic cytotoxic T-cell lymphoma. Genes Chromosomes Cancer 2018; 57:622-629. [DOI: 10.1002/gcc.22673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/27/2022] Open
Affiliation(s)
- Daniele Fanoni
- Dipartimento di Fisiopatologia medico-chirurgica e dei trapianti; Universitá degli Studi di Milano; Milan Italy
| | - Laura Corti
- Department of Dermatology; Fondazione IRCCS Ca’ Granda - Ospedale Maggiore Policlinico; Milan Italy
| | - Silvia Alberti-Violetti
- Department of Dermatology; Fondazione IRCCS Ca’ Granda - Ospedale Maggiore Policlinico; Milan Italy
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale; Universitá degli Studi di Milano; Milan Italy
| | - Cornelis P. Tensen
- Department of Dermatology; Leiden University Medical Centre (LUMC); Leiden The Netherlands
| | - Luigia Venegoni
- Dipartimento di Fisiopatologia medico-chirurgica e dei trapianti; Universitá degli Studi di Milano; Milan Italy
| | - Maarten Vermeer
- Department of Dermatology; Leiden University Medical Centre (LUMC); Leiden The Netherlands
| | - Rein Willemze
- Department of Dermatology; Leiden University Medical Centre (LUMC); Leiden The Netherlands
| | - Emilio Berti
- Dipartimento di Fisiopatologia medico-chirurgica e dei trapianti; Universitá degli Studi di Milano; Milan Italy
- Department of Dermatology; Fondazione IRCCS Ca’ Granda - Ospedale Maggiore Policlinico; Milan Italy
| |
Collapse
|
49
|
Stadler R, Stranzenbach R. Molecular pathogenesis of cutaneous lymphomas. Exp Dermatol 2018; 27:1078-1083. [DOI: 10.1111/exd.13701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Rudolf Stadler
- University Clinic for Dermatology, Venerology, Allergology and Phlebology; Johannes Wesling Medical Centre; UKRUB; University of Bochum; Minden Germany
| | - René Stranzenbach
- University Clinic for Dermatology, Venerology, Allergology and Phlebology; Johannes Wesling Medical Centre; UKRUB; University of Bochum; Minden Germany
| |
Collapse
|
50
|
Gorenkova LG, Kravchenko SK, Kovrigina AM, Kolomeitsev OA. Successful sequential immune epigenetic therapy of erythrodermic mycosis fungoidesessful sequential immune epigenetic therapy with the resistant course of erythrodermic mycosis fungoides. VESTNIK DERMATOLOGII I VENEROLOGII 2018. [DOI: 10.25208/0042-4609-2018-94-3-77-84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Mucosis fungoidea (МF) belongs to the class of epidermotropic T-cell lymphomas. MF is represented by over 10 sub-types only in terms of its clinical manifestations, with one of them being erythrodermic MF (EMF). This disease is characterized by diverse symptomatology in the form of erythroderma and intense skin itch, aggressive сlinical course and unfavorable prognosis. The disease prognosis also correlates with age, previous history of long-term systemic gluco-corticosteroid treatment (GCS), increased activity of lactate dehydrogenase (LDH) and hypereosinophilia. The choice of MF treatment is determined by the disease stage and somatic status of the patient. In EMF, a therapy combining various effective preparations and taking into account the specifics of the given case is required. Extracorporeal photopheresis (ECP) is frequently an approach of choice; however, it has demonstrated the highest efficacy in Sezary disease or in EFM associated with leucemization. Application of new pharmaceuticals (monoclonal antibodies, epigenetic agents) in combination or in sequence with immune therapy is a promising direction, particularly for treating patients older than 75 years. In this paper, we describe the clinical case of an elderly patient suffering from EMF without peripheral blood leukemia with multimodal factors of unfavorable prognosis, such as age, increased lactate dehy drogenase activity, history of prolonged inefficient treatment with gluco-cortecosteroid preparations and eosinophilia. A long-term positive response to the treatment using sequential immune epigenetic therapy has not been achieved, although the treatment tolerability and the patient's life quality were satisfactory.
Collapse
|