1
|
Leung E, Pryma C, Murphy S, Harrison R, Peterson E, Tsang PWK, Varghese J, You XJ, Slack GW, Skinnider BF, Ng T, Young S, Burrell S, Stubbins R, Lim H, Carruthers M, Dutz J, Diamond EL, Chen LYC. Rosai-Dorfman-Destombes disease in adults: a single center experience. Ann Hematol 2024; 103:4467-4476. [PMID: 39331156 DOI: 10.1007/s00277-024-06019-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Recent advances in Rosai-Dorfman-Destombes disease (RDD), notably molecular testing, targeted therapy, and PET-CT imaging, hold promise for better recognition and improved outcomes. This study presents patients diagnosed and treated in a "real world" setting, where navigating limited resources must be considered. This retrospective single-center review includes 15 adult patients diagnosed with RDD at Vancouver General Hospital between November 2015 and October 2023. The cohort comprised five males and ten females with a median age 53 years (range 19-80 years). All 15 patients had extra-nodal disease; 11 patients exclusively had extra-nodal disease, and four patients also had lymph node involvement. Seven patients had tissue next-generation sequencing, identifying MAP2K1 mutations in four cases and a KRAS p.K117N mutation in one case that was treated with targeted therapy using trametinib. PET-CT was used for disease staging in four cases. Six patients with refractory disease tolerated lenalidomide and dexamethasone without significant toxicity; three patients achieved complete response, and three had partial response. This study highlights RDD's diverse extra-nodal manifestations. Lenalidomide combined with dexamethasone is an effective and well-tolerated treatment option for select patients, especially those with refractory disease. Broad utilization of NGS and PET-CT can positively influence management decisions.
Collapse
Affiliation(s)
- Emily Leung
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Collin Pryma
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Murphy
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Rebecca Harrison
- Department of Neurology, University of British Columbia, Vancouver, BC, Canada
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Erica Peterson
- Division of Hematology, University of British Columbia, Vancouver, BC, Canada
| | - Peter W K Tsang
- Division of Hematology, University of British Columbia, Vancouver, BC, Canada
| | - Julia Varghese
- Division of Hematology, University of British Columbia, Vancouver, BC, Canada
| | | | - Graham W Slack
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Brian F Skinnider
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tony Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Sean Young
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Cancer Genetics and Genomics Laboratory, BC Cancer Agency, Vancouver, BC, Canada
| | - Steven Burrell
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
| | - Ryan Stubbins
- Division of Hematology, University of British Columbia, Vancouver, BC, Canada
| | - Howard Lim
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | | | - Jan Dutz
- Department of Dermatology and Skin Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eli L Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Luke Y C Chen
- Division of Hematology, University of British Columbia, Vancouver, BC, Canada.
- Division of Hematology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
2
|
Nguyen LT, Pham GH, Vu PT, Yi HG. Favorable outcome of a histiocytic sarcoma patient treated with immune checkpoint inhibitor: a case report. Ann Med Surg (Lond) 2023; 85:6274-6278. [PMID: 38098600 PMCID: PMC10718375 DOI: 10.1097/ms9.0000000000001446] [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: 07/25/2023] [Accepted: 10/18/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction and Importance Histiocytic sarcoma (HS) is an extremely rare malignancy in which there has been no standard treatment approach. Some preclinical studies have provided rationales for the application of immunotherapy in advanced HS. Case Presentation The authors reported a case of a 61-year-old patient with metastatic HS who had a rapid progression on ifosfamide, carboplatin, and etoposide chemotherapy. The authors performed PD-L1 testing, which showed a strong positivity in 90% of tumor cells. The patient was then treated with pembrolizumab 200 mg every 3 weeks. He refused palliative radiotherapy. A dramatic response in all sites was recorded on the PET-CT scan after three cycles. He was maintained on pembrolizumab, reaching over 30 months without disease progression. Clinical Discussion Recent molecular data suggests there could be a role of immunotherapy in HS. In our patient, the disease was refractory to chemotherapy and pembrolizumab has been given based on the strong PD-L1 expression. Response to immunotherapy has also been recorded in several cases with malignant histiocytic neoplasm. Conclusion Immunotherapy might bring sustained disease remission in PD-L1 high expression HS and further studies evaluating the role of immune checkpoint inhibitor in this disease are warranted.
Collapse
Affiliation(s)
- Long Thanh Nguyen
- Department of Medical Oncology No. 6, Vietnam National Cancer Hospital
| | | | - Phuong Thi Vu
- Department of Laboratory, Vinmec Times City International General Hospital
| | - Hyeon Gyu Yi
- Department of Oncology and Hematology
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
| |
Collapse
|
3
|
Gaulin C, Craig F, Fortin Ensign S, Robetorye R, Gibson S, Kelemen K, Khera N, Leis J, Tsang M, Rosenthal A, Munoz J, Palmer J, Hilal T. Allogeneic hematopoietic stem cell transplantation in adults with dendritic/histiocytic neoplasms. Leuk Lymphoma 2023; 64:2347-2350. [PMID: 37680001 DOI: 10.1080/10428194.2023.2255707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Affiliation(s)
- Charles Gaulin
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Fiona Craig
- Department of Laboratory Medicine and Pathology, Division of Hematopathology, Mayo Clinic, Phoenix, AZ, USA
| | - Shannon Fortin Ensign
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Ryan Robetorye
- Department of Laboratory Medicine and Pathology, Division of Hematopathology, Mayo Clinic, Phoenix, AZ, USA
| | - Sarah Gibson
- Department of Laboratory Medicine and Pathology, Division of Hematopathology, Mayo Clinic, Phoenix, AZ, USA
| | - Katalin Kelemen
- Department of Laboratory Medicine and Pathology, Division of Hematopathology, Mayo Clinic, Phoenix, AZ, USA
| | - Nandita Khera
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Jose Leis
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Mazie Tsang
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Allison Rosenthal
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Javier Munoz
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Jeanne Palmer
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Talal Hilal
- Department of Medicine, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
4
|
Li Y, Wang X, Hou X, Ma X. Could Inhibiting the DNA Damage Repair Checkpoint Rescue Immune-Checkpoint-Inhibitor-Resistant Endometrial Cancer? J Clin Med 2023; 12:jcm12083014. [PMID: 37109350 PMCID: PMC10144486 DOI: 10.3390/jcm12083014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/23/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Endometrial cancer (EC) is increasingly undermining female health worldwide, with poor survival rates for advanced or recurrent/metastatic diseases. The application of immune checkpoint inhibitors (ICIs) has opened a window of opportunity for patients with first-line therapy failure. However, there is a subset of patients with endometrial cancer who remain insensitive to immunotherapy alone. Therefore, it is necessary to develop new therapeutic agents and further explore reliable combinational strategies to optimize the efficacy of immunotherapy. DNA damage repair (DDR) inhibitors as novel targeted drugs are able to generate genomic toxicity and induce cell death in solid tumors, including EC. Recently, growing evidence has demonstrated the DDR pathway modulates innate and adaptive immunity in tumors. In this review, we concentrate on the exploration of the intrinsic correlation between DDR pathways, especially the ATM-CHK2-P53 pathway and the ATR-CHK1-WEE1 pathway, and oncologic immune response, as well as the feasibility of adding DDR inhibitors to ICIs for the treatment of patients with advanced or recurrent/metastatic EC. We hope that this review will offer some beneficial references to the investigation of immunotherapy and provide a reasonable basis for "double-checkpoint inhibition" in EC.
Collapse
Affiliation(s)
- Yinuo Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Hou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangyi Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
5
|
Clinical Significance of Serum Biomarkers in Stage IV Non-Small-Cell Lung Cancer Treated with PD-1 Inhibitors: LIPI Score, NLR, dNLR, LMR, and PAB. DISEASE MARKERS 2022; 2022:7137357. [PMID: 35945957 PMCID: PMC9357262 DOI: 10.1155/2022/7137357] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/08/2022] [Indexed: 12/24/2022]
Abstract
Background To assess the prognostic value of pretreatment serum biomarkers in stage IV non-small-cell lung cancer (NSCLC) patients treated with PD-1 (programmed cell death protein 1) inhibitors and their value as a predictor of benefit. Methods We performed a retrospective study including patients with stage IV NSCLC who were treated with anti-PD-1 drugs in first or advanced lines of therapy in the Affiliated Tumor Hospital of Nantong University. Serum biomarkers such as NLR, dNLR, LMR, PAB, ALB, and LIPI scores were calculated and analyzed in detail. Results A total of 85 patients with stage IV NSCLC treated with PD-1 inhibitors in the first or advanced lines of therapy were included in this subject. According to the tumor response of PD-1-based treatment, ORR was 42.4% (36/85) and DCR was 68.2% (58/85). The median OS and PFS were 20.0 months and 7.0 months, respectively. The ROC curves showed that the serum biomarkers of NLR, dNLR, LDH, LMR, PAB, and ALB were significantly associated with overall survival and helped to determine the cut-off value. The multivariate Cox proportional hazard analyses for stage IV NSCLC patients treated with PD-1 inhibitors indicated that dNLR (P < 0.001) and ALB (P = 0.033) were independent prognostic indicators of PFS, while liver metastasis (P = 0.01), NLR (P = 0.01), dNLR (P = 0.001), and LMR (P = 0.006) were independent prognostic indicators of OS. Moreover, patients of the good LIPI group showed prolonged PFS and OS than those with intermediate/poor LIPI score (P < 0.001 and P = 0.006, respectively). Conclusions Pretreatment dNLR is an independent prognostic indicator of both PFS and OS in stage IV NSCLC patients treated with PD-1 inhibitors. Pretreatment LIPI, combining dNLR > 3 and LDH>ULN, was correlated with worse outcome for stage IV NSCLC patients treated with ICI. High NLR, high dNLR, low LMR, and low ALB at baseline might be useful as an early predictive biomarker of benefit.
Collapse
|
6
|
Furui Y, Kurata T, Komori K, Uchida E, Miyairi Y, Chiba A, Ogiso Y, Sakashita K. A case of recurrent refractory cervical primary histiocytic sarcoma treated with pembrolizumab. Int Cancer Conf J 2022; 11:280-285. [DOI: 10.1007/s13691-022-00562-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022] Open
|
7
|
Srivastava RM, Purohit TA, Chan TA. Diverse Neoantigens and the Development of Cancer Therapies. Semin Radiat Oncol 2021; 30:113-128. [PMID: 32381291 DOI: 10.1016/j.semradonc.2019.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer is the manifestation of uncontrolled cellular growth and immune escape mechanisms. Unrestrained tumor growth can be associated with incidental errors in the genome during replication and genotoxic agents can alter the structure and sequence of our DNA. Among all genetic aberrations in cancer, only limited number of mutations can produce immunogenic antigens which have the potential to bind human leukocyte antigen class I or human leukocyte antigen class II, and help activate the adaptive immune system. These neoantigens can be recognized by CD8+ and CD4+ neoantigen-specific T lymphocytes. Recently, several immune checkpoint targeting drugs have been approved for clinical use. Primarily, these drugs expand and facilitate the cytotoxic activity of neoantigen-specific T cells to eradicate tumors. Differential drug response across cancers could be attributed, at least in part, to differences in the 'tumor antigen landscape' and 'antigen presentation pathway' in patients. Although tumor mutational burden correlates with response to immune checkpoint inhibitors in many cancer types and has evolved as a broad biomarker, a comprehensive understanding of the neoantigen landscape and the function of cognate T cell responses is lacking and is needed for improved patient selection criteria and neoantigen vaccine design. Here, we review cancer neoantigens, their implications for antitumor responses, the dynamics of neoantigen-specific T cells, and the advancement of neoantigen-based therapy in proposed clinical trials.
Collapse
Affiliation(s)
- Raghvendra M Srivastava
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tanaya A Purohit
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Timothy A Chan
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
8
|
Montalvo N, Lara-Endara J, Redrobán L, Leiva M, Armijos C, Russo L. Primary splenic histiocytic sarcoma associated with hemophagocytic lymphohistiocytosis: A case report and review of literature of next-generation sequencing involving FLT3, NOTCH2, and KMT2A mutations. Cancer Rep (Hoboken) 2021; 5:e1496. [PMID: 34292677 PMCID: PMC9124500 DOI: 10.1002/cnr2.1496] [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: 04/07/2021] [Revised: 06/01/2021] [Accepted: 06/17/2021] [Indexed: 11/25/2022] Open
Abstract
Background Histiocytic sarcoma is a very rare monocyte/macrophage‐derived hematopoietic system tumor with a poor prognosis whose diagnosis is pathologically challenging due to its extreme rarity and histological overlap with various mimicking entities in which histiocytes also predominate. Case We report the case of a 33‐year‐old male patient with hemophagocytic lymphohistiocytosis, purpuric syndrome, and significant splenomegaly. The patient underwent splenectomy; subsequent macroscopic examination revealed a spleen weighing 2065 grams with hyperemic red pulp and multiple infarcts at the periphery. The histological and immunohistochemical study established a diagnosis of primary splenic histiocytic sarcoma with frequent hemophagocytosis. Next‐generation sequencing demonstrated mutations in FLT3, NOTCH2, and KMT2A, microsatellite stability, and a tumor mutational burden of 2 mut/Mb. The patient's condition deteriorated clinically from the appearance of the first symptoms and he died 6 months later from multi‐organ failure. Conclusion Primary splenic histiocytic sarcoma is one of the rarest tumors of the hematopoietic system. We report the first case with mutations in FLT3, NOTCH2, and KMT2A, and associated hemophagocytic lymphohistiocytosis.
Collapse
Affiliation(s)
- Nelson Montalvo
- Facultad de Ciencias Médicas de la Salud y la Vida, Escuela de Medicina, Departamento de Docencia e Investigación, Universidad Internacional del Ecuador, Av. Simón Bolívar y Jorge Fernández, Quito, Ecuador.,Servicio de Patología, Hospital Metropolitano, Av. Mariana de Jesús s/n y Nicolás Arteta, Quito, Ecuador
| | - Jorge Lara-Endara
- Facultad de Ciencias Médicas de la Salud y la Vida, Escuela de Medicina, Departamento de Docencia e Investigación, Universidad Internacional del Ecuador, Av. Simón Bolívar y Jorge Fernández, Quito, Ecuador
| | - Ligia Redrobán
- Servicio de Patología, Hospital Metropolitano, Av. Mariana de Jesús s/n y Nicolás Arteta, Quito, Ecuador
| | - María Leiva
- Servicio de Hematología, Hospital Metropolitano, Av. Mariana de Jesús s/n y Nicolás Arteta, Quito, Ecuador
| | - Christian Armijos
- Facultad de Ciencias Médicas de la Salud y la Vida, Escuela de Medicina, Departamento de Docencia e Investigación, Universidad Internacional del Ecuador, Av. Simón Bolívar y Jorge Fernández, Quito, Ecuador.,Servicio de Radiología, Hospital Metropolitano, Av. Mariana de Jesús s/n y Nicolás Arteta, Quito, Ecuador
| | - Leonardo Russo
- Facultad de Ciencias Médicas de la Salud y la Vida, Escuela de Medicina, Departamento de Docencia e Investigación, Universidad Internacional del Ecuador, Av. Simón Bolívar y Jorge Fernández, Quito, Ecuador
| |
Collapse
|
9
|
Cao W, Ma X, Fischer JV, Sun C, Kong B, Zhang Q. Immunotherapy in endometrial cancer: rationale, practice and perspectives. Biomark Res 2021; 9:49. [PMID: 34134781 PMCID: PMC8207707 DOI: 10.1186/s40364-021-00301-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor immunotherapy has attracted more and more attention nowadays, and multiple clinical trials have confirmed its effect in a variety of solid tumors. Immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell transfer (ACT), and lymphocyte-promoting cytokines are the main immunotherapy methods. Endometrial cancer (EC) is one of the most frequent tumors in women and the prognosis of recurrent or metastatic EC is poor. Since molecular classification has been applied to EC, immunotherapy for different EC subtypes (especially POLE and MSI-H) has gradually attracted attention. In this review, we focus on the expression and molecular basis of the main biomarkers in the immunotherapy of EC firstly, as well as their clinical application significance and limitations. Blocking tumor immune checkpoints is one of the most effective strategies for cancer treatment in recent years, and has now become the focus in the field of tumor research and treatment. We summarized clinical date of planned and ongoing clinical trials and introduced other common immunotherapy methods in EC, such as cancer vaccine and ACT. Hormone aberrations, metabolic syndrome (MetS) and p53 mutant and that affect the immunotherapy of endometrial cancer will also be discussed in this review.
Collapse
Affiliation(s)
- Wenyu Cao
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Xinyue Ma
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Jean Victoria Fischer
- Department of Pathology, Northwestern Medicine, Gynecologic Pathology Fellow, Chicago, Illinois, USA
| | - Chenggong Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China. .,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China.
| |
Collapse
|
10
|
Erdheim-Chester disease: consensus recommendations for evaluation, diagnosis, and treatment in the molecular era. Blood 2021; 135:1929-1945. [PMID: 32187362 DOI: 10.1182/blood.2019003507] [Citation(s) in RCA: 196] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/04/2020] [Indexed: 01/18/2023] Open
Abstract
Erdheim-Chester disease (ECD) is a rare histiocytosis that was recently recognized as a neoplastic disorder owing to the discovery of recurrent activating MAPK (RAS-RAF-MEK-ERK) pathway mutations. Typical findings of ECD include central diabetes insipidus, restrictive pericarditis, perinephric fibrosis, and sclerotic bone lesions. The histopathologic diagnosis of ECD is often challenging due to nonspecific inflammatory and fibrotic findings on histopathologic review of tissue specimens. Additionally, the association of ECD with unusual tissue tropism and an insidious onset often results in diagnostic errors and delays. Most patients with ECD require treatment, except for a minority of patients with minimally symptomatic single-organ disease. The first ECD consensus guidelines were published in 2014 on behalf of the physicians and researchers within the Erdheim-Chester Disease Global Alliance. With the recent molecular discoveries and the approval of the first targeted therapy (vemurafenib) for BRAF-V600-mutant ECD, there is a need for updated clinical practice guidelines to optimize the diagnosis and treatment of this disease. This document presents consensus recommendations that resulted from the International Medical Symposia on ECD in 2017 and 2019. Herein, we include the guidelines for the clinical, laboratory, histologic, and radiographic evaluation of ECD patients along with treatment recommendations based on our clinical experience and review of literature in the molecular era.
Collapse
|
11
|
Hédan B, Rault M, Abadie J, Ulvé R, Botherel N, Devauchelle P, Copie-Bergman C, Cadieu E, Parrens M, Alten J, Zalcman EL, Cario G, Damaj G, Mokhtari K, Le Loarer F, Coulomb-Lhermine A, Derrien T, Hitte C, Bachelot L, Breen M, Gilot D, Blay JY, Donadieu J, André C. PTPN11 mutations in canine and human disseminated histiocytic sarcoma. Int J Cancer 2020; 147:1657-1665. [PMID: 32212266 DOI: 10.1002/ijc.32991] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 01/20/2023]
Abstract
In humans, histiocytic sarcoma (HS) is an aggressive cancer involving histiocytes. Its rarity and heterogeneity explain that treatment remains a challenge. Sharing high clinical and histopathological similarities with human HS, the canine HS is conversely frequent in specific breeds and thus constitutes a unique spontaneous model for human HS to decipher the genetic bases and to explore therapeutic options. We identified sequence alterations in the MAPK pathway in at least 63.9% (71/111) of HS cases with mutually exclusive BRAF (0.9%; 1/111), KRAS (7.2%; 8/111) and PTPN11 (56.75%; 63/111) mutations concentrated at hotspots common to human cancers. Recurrent PTPN11 mutations are associated to visceral disseminated HS subtype in dogs, the most aggressive clinical presentation. We then identified PTPN11 mutations in 3/19 (15.7%) human HS patients. Thus, we propose PTPN11 mutations as key events for a specific subset of human and canine HS: the visceral disseminated form. Finally, by testing drugs targeting the MAPK pathway in eight canine HS cell lines, we identified a better anti-proliferation activity of MEK inhibitors than PTPN11 inhibitors in canine HS neoplastic cells. In combination, these results illustrate the relevance of naturally affected dogs in deciphering genetic mechanisms and selecting efficient targeted therapies for such rare and aggressive cancers in humans.
Collapse
Affiliation(s)
- Benoit Hédan
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Mélanie Rault
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Jérôme Abadie
- Department of Biology, Pathology and Food Sciences, Oniris, Laboniris, Nantes, France
| | - Ronan Ulvé
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Nadine Botherel
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | | | - Christiane Copie-Bergman
- Assistance Publique-Hôpitaux de Paris, Département de Pathologie, Groupe Henri-Mondor Albert-Chenevier, Créteil, France.,INSERM U955, Équipe 9, Faculté de Médecine, Université Paris Est Créteil, Créteil, France
| | - Edouard Cadieu
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Marie Parrens
- Department of Pathology, CHU de Bordeaux, Hôpital du Haut Lévêque, INSERM U1035, Université de Bordeaux, Bordeaux, France
| | - Julia Alten
- Pediatric Oncology/Hematology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Emmanuelle L Zalcman
- Department of Neuropathology, GHU Paris Psychiatrie Neurosciences, Sainte-Anne Hospital, Paris, France
| | - Gunnar Cario
- Pediatric Oncology/Hematology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Gandhi Damaj
- Haemalology Institute, CHU de Caen and Centre François Baclesse, Caen, France
| | - Karima Mokhtari
- Sorbonne University, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neuropathologie-Escourolle, Paris, France
| | | | | | - Thomas Derrien
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Christophe Hitte
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Laura Bachelot
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, CVM Research Building, Raleigh, NC, USA
| | - David Gilot
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| | - Jean Y Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Jean Donadieu
- Department of Haematology, APHP, Trousseau Hospital, Paris, France
| | - Catherine André
- Faculty of Medicine, CNRS-University of Rennes 1, UMR6290, Institute of Genetics and Development of Rennes, SFR Biosit, Rennes, France
| |
Collapse
|
12
|
Goyal G, Ravindran A, Young JR, Shah MV, Bennani NN, Patnaik MM, Nowakowski GS, Thanarajasingam G, Habermann TM, Vassallo R, Sher T, Parikh SA, Rech KL, Go RS. Clinicopathological features, treatment approaches, and outcomes in Rosai-Dorfman disease. Haematologica 2020; 105:348-357. [PMID: 31004029 PMCID: PMC7012468 DOI: 10.3324/haematol.2019.219626] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/18/2019] [Indexed: 12/11/2022] Open
Abstract
Rosai-Dorfman disease is a rare subtype of non-Langerhans cell histiocytosis. With the last major report published in 1990, there is a paucity of contemporary data on this disease. Our objective was to report the clinicopathological features, treatments and outcomes of patients seen at a tertiary referral center. Sixty-four patients with histopathological diagnosis of Rosai-Dorfman disease were identified from 1994 to 2017 (median age 50 years; range, 2-79). The median duration from symptom onset to diagnosis was seven months (range, 0-128), which was also reflected in the number of biopsies required to establish the diagnosis (median 2; range, 1-6). The most common presentation was subcutaneous masses (40%). Of the 64 patients, 8% had classical (nodal only) and 92% had extra-nodal disease (67% extra-nodal only). The most common organs involved were skin and subcutaneous tissue (52%), followed by lymph nodes (33%). Three patients had an overlap with Erdheim-Chester disease, which had not been described before. Two of these were found to have MAP2K1 mutations. Commonly utilized first line treatments were surgical excision (38%) and systemic corticosteroids (27%). Corticosteroids led to a response in 56% of the cases. Of those treated initially, 15 (30%) patients developed recurrent disease. The most commonly used systemic agent was cladribine (n=6), with 67% overall response rate. Our study demonstrates that Rosai-Dorfman disease has diverse clinical manifestations and outcomes. While this disease has been historically considered a benign entity, a subset of patients endures an aggressive course necessitating the use of systemic therapies.
Collapse
Affiliation(s)
- Gaurav Goyal
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | | | | | - Taimur Sher
- Division of Hematology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Karen L Rech
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Ronald S Go
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
13
|
Kemps PG, Zondag TC, Steenwijk EC, Andriessen Q, Borst J, Vloemans S, Roelen DL, Voortman LM, Verdijk RM, van Noesel CJM, Cleven AHG, Hawkins C, Lang V, de Ru AH, Janssen GMC, Haasnoot GW, Franken KLMC, van Eijk R, Solleveld-Westerink N, van Wezel T, Egeler RM, Beishuizen A, van Laar JAM, Abla O, van den Bos C, van Veelen PA, van Halteren AGS. Apparent Lack of BRAF V600E Derived HLA Class I Presented Neoantigens Hampers Neoplastic Cell Targeting by CD8 + T Cells in Langerhans Cell Histiocytosis. Front Immunol 2020; 10:3045. [PMID: 31998317 PMCID: PMC6967030 DOI: 10.3389/fimmu.2019.03045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Langerhans Cell Histiocytosis (LCH) is a neoplastic disorder of hematopoietic origin characterized by inflammatory lesions containing clonal histiocytes (LCH-cells) intermixed with various immune cells, including T cells. In 50-60% of LCH-patients, the somatic BRAF V600E driver mutation, which is common in many cancers, is detected in these LCH-cells in an otherwise quiet genomic landscape. Non-synonymous mutations like BRAF V600E can be a source of neoantigens capable of eliciting effective antitumor CD8+ T cell responses. This requires neopeptides to be stably presented by Human Leukocyte Antigen (HLA) class I molecules and sufficient numbers of CD8+ T cells at tumor sites. Here, we demonstrate substantial heterogeneity in CD8+ T cell density in n = 101 LCH-lesions, with BRAF V600E mutated lesions displaying significantly lower CD8+ T cell:CD1a+ LCH-cell ratios (p = 0.01) than BRAF wildtype lesions. Because LCH-lesional CD8+ T cell density had no significant impact on event-free survival, we investigated whether the intracellularly expressed BRAF V600E protein is degraded into neopeptides that are naturally processed and presented by cell surface HLA class I molecules. Epitope prediction tools revealed a single HLA class I binding BRAF V600E derived neopeptide (KIGDFGLATEK), which indeed displayed strong to intermediate binding capacity to HLA-A*03:01 and HLA-A*11:01 in an in vitro peptide-HLA binding assay. Mass spectrometry-based targeted peptidomics was used to investigate the presence of this neopeptide in HLA class I presented peptides isolated from several BRAF V600E expressing cell lines with various HLA genotypes. While the HLA-A*02:01 binding BRAF wildtype peptide KIGDFGLATV was traced in peptides isolated from all five cell lines expressing this HLA subtype, KIGDFGLATEK was not detected in the HLA class I peptidomes of two distinct BRAF V600E transduced cell lines with confirmed expression of HLA-A*03:01 or HLA-A*11:01. These data indicate that the in silico predicted HLA class I binding and proteasome-generated neopeptides derived from the BRAF V600E protein are not presented by HLA class I molecules. Given that the BRAF V600E mutation is highly prevalent in chemotherapy refractory LCH-patients who may qualify for immunotherapy, this study therefore questions the efficacy of immune checkpoint inhibitor therapy in LCH.
Collapse
Affiliation(s)
- Paul G Kemps
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Timo C Zondag
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eline C Steenwijk
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Quirine Andriessen
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Jelske Borst
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Sandra Vloemans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Dave L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Lenard M Voortman
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Robert M Verdijk
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carel J M van Noesel
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Cynthia Hawkins
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Veronica Lang
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Arnoud H de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - George M C Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Geert W Haasnoot
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Kees L M C Franken
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Ronald van Eijk
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - R Maarten Egeler
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands.,Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Auke Beishuizen
- Department of Pediatric Oncology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Jan A M van Laar
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Oussama Abla
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Cor van den Bos
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Astrid G S van Halteren
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
14
|
Roloff GW, Baron JI, Neppalli VT, Sait S, Griffiths EA. Next-Generation Sequencing Delineates Clonal Origins and Informs Therapeutic Strategies in Acute Lymphoblastic Leukemia and Histiocytic Sarcoma. JCO Precis Oncol 2019; 3:1-8. [PMID: 35100717 DOI: 10.1200/po.19.00126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
| | | | | | - Sheila Sait
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | |
Collapse
|
15
|
Goyal G, Young JR, Koster MJ, Tobin WO, Vassallo R, Ryu JH, Davidge-Pitts CJ, Hurtado MD, Ravindran A, Sartori Valinotti JC, Bennani NN, Shah MV, Rech KL, Go RS. The Mayo Clinic Histiocytosis Working Group Consensus Statement for the Diagnosis and Evaluation of Adult Patients With Histiocytic Neoplasms: Erdheim-Chester Disease, Langerhans Cell Histiocytosis, and Rosai-Dorfman Disease. Mayo Clin Proc 2019; 94:2054-2071. [PMID: 31472931 DOI: 10.1016/j.mayocp.2019.02.023] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/14/2019] [Accepted: 02/22/2019] [Indexed: 12/25/2022]
Abstract
Histiocytic neoplasms, a rare and heterogeneous group of disorders, primarily include Erdheim-Chester disease, Langerhans cell histiocytosis, and Rosai-Dorfman disease. Due to their diverse clinical manifestations, the greatest challenge posed by these neoplasms is the establishment of a diagnosis, which often leads to a delay in institution of appropriate therapy. Recent insights into their genomic architecture demonstrating mitogen-activated protein kinase/extracellular signal-regulated kinase pathway mutations have now enabled potential treatment with targeted therapies in most patients. This consensus statement represents a joint document from a multidisciplinary group of physicians at Mayo Clinic who specialize in the management of adult histiocytic neoplasms. It consists of evidence- and consensus-based recommendations on when to suspect these neoplasms and what tests to order for the diagnosis and initial evaluation. In addition, it also describes the histopathologic and individual organ manifestations of these neoplasms to help the clinicians in identifying their key features. With uniform guidelines that aid in identifying these neoplasms, we hope to improve the awareness that may lead to their timely and correct diagnosis.
Collapse
Affiliation(s)
- Gaurav Goyal
- Division of Hematology, Mayo Clinic, Rochester, MN.
| | | | | | | | - Robert Vassallo
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Jay H Ryu
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | | | - Maria D Hurtado
- Division of Endocrinology, Diabetes, and Nutrition, Mayo Clinic, Rochester, MN
| | | | | | | | | | - Karen L Rech
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Ronald S Go
- Division of Hematology, Mayo Clinic, Rochester, MN.
| | | |
Collapse
|
16
|
Satou A, Bennani NN, Feldman AL. Update on the classification of T-cell lymphomas, Hodgkin lymphomas, and histiocytic/dendritic cell neoplasms. Expert Rev Hematol 2019; 12:833-843. [PMID: 31365276 DOI: 10.1080/17474086.2019.1647777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: The classification of lymphomas is based on the postulated normal counterparts of lymphoid neoplasms and currently constitutes over 100 definite or provisional entities. As this number of entities implies, lymphomas show marked pathological, genetic, and clinical heterogeneity. Recent molecular findings have significantly advanced our understanding of lymphomas. Areas covered: The World Health Organization (WHO) classification of lymphoid neoplasms was updated in 2017. The present review summarizes the new findings that have been gained in the areas of mature T-cell neoplasms, Hodgkin lymphomas, and histiocytic/dendritic cell neoplasms since the publication of the 2017 WHO classification. Expert opinion: Although formal revisions to the WHO classification are published only periodically, our understanding of the pathologic, genetic, and clinical features of lymphoid neoplasms is constantly evolving, particularly in the age of -omics technologies and targeted therapeutics. Even in the relatively short time since the publication of the 2017 WHO classification, many significant findings have been identified in the entities covered in this review.
Collapse
Affiliation(s)
- Akira Satou
- Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester , MN , USA.,Department of Surgical Pathology, Aichi Medical University Hospital , Nagakute , Aichi , Japan
| | - N Nora Bennani
- Division of Hematology, Mayo Clinic , Rochester , MN , USA
| | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|