1
|
Lowe EJ, Woessmann W. Anaplastic large cell lymphoma in children and adolescents. Br J Haematol 2025. [PMID: 40351161 DOI: 10.1111/bjh.20154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Anaplastic lymphoma kinase (ALK)-positive anaplastic large-cell lymphoma (ALCL) accounts for >95% of ALCL cases in children and adolescents. The first description of ALCL as a CD30-positive lymphoma in 1985 was followed by the detection of chromosomal translocations involving the ALK gene at chromosome 2p23. The pathogenesis of ALK-positive ALCL is based on signalling from the constitutive active ALK kinase. The clinical characteristics, therapy regimens and outcome data were reported in the 1990s and 2000s. Different chemotherapy regimens led to astonishingly similar long-term event-free survival rates of 70%, independent of the drugs, doses and duration of therapy. Additionally, patients with relapsed ALCL are treated successfully with very different re-induction and consolidation approaches leading to an overall survival approaching 95%. In the 2010s, minimal disseminated and residual disease, histological subtype and antibody titres against ALK were reported as significant independent prognostic factors. Over the last 15 years, targeted therapies (brentuximab vedotin, ALK inhibitors) have demonstrated high efficacy with low toxicity. The future of ALK-positive ALCL is to define the role of targeted therapies by developing a 'chemotherapy-free' approach for patients with standard-risk ALCL and an integrated approach to cure patients with high-risk ALCL.
Collapse
Affiliation(s)
- Eric J Lowe
- Department of Pediatrics, Eastern Virginia Medical School at Old Dominion University, Norfolk, Virginia, USA
| | - Wilhelm Woessmann
- Pediatric Hematology and Oncology and NHL-BFM Study Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
Voena C, Ambrogio C, Iannelli F, Chiarle R. ALK in cancer: from function to therapeutic targeting. Nat Rev Cancer 2025; 25:359-378. [PMID: 40055571 DOI: 10.1038/s41568-025-00797-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 05/01/2025]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that acts as an oncogenic driver in solid and haematological malignancies in both children and adults. Although ALK-expressing (ALK+) tumours show strong initial responses to the series of ALK inhibitors currently available, many patients will develop resistance. In this Review, we discuss recent advances in ALK oncogenic signalling, together with existing and promising new modalities to treat ALK-driven tumours, including currently approved ALK-directed therapies, namely tyrosine kinase inhibitors, and novel approaches such as ALK-specific immune therapies. Although ALK inhibitors have changed the management and clinical history of ALK+ tumours, they are still insufficient to cure most of the patients. Therefore, more effort is needed to further improve outcomes and prevent the tumour resistance, recurrence and metastatic spread that many patients with ALK+ tumours experience. Here, we outline how a multipronged approach directed against ALK and other essential pathways that sustain the persistence of ALK+ tumours, together with potent or specific immunotherapies, could achieve this goal. We envision that the lessons learned from treating ALK+ tumours in the clinic could ultimately accelerate the implementation of innovative combination therapies to treat tumours driven by other tyrosine kinases or oncogenes with similar properties.
Collapse
Affiliation(s)
- Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Fabio Iannelli
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy.
- Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Marks LJ, Ritter V, Agrusa JE, Kamdar KY, Rivers J, Gardner R, Ehrhardt MJ, Devine KJ, Phillips CA, Reilly A, August K, Weinstein J, Satwani P, Forlenza CJ, Smith CM, Greer C, Afify Z, Lin CH, Belsky JA, Ding H, Hoogstra D, Toner K, Link MP, Schultz LM, Lowe EJ, Aftandilian C. Pediatric relapsed/refractory ALK-positive anaplastic large cell lymphoma treatment and outcomes in the targeted-drug era. Blood Adv 2025; 9:1356-1365. [PMID: 39841960 PMCID: PMC11954107 DOI: 10.1182/bloodadvances.2024014745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/24/2025] Open
Abstract
ABSTRACT Treatment options for patients with relapsed or refractory (R/R) anaplastic large cell lymphoma (ALCL) have increased in the era of targeted therapies such as brentuximab vedotin (BV) and anaplastic lymphoma kinase (ALK) inhibitors. However, there is no standard treatment and published data evaluating their use are limited. The goal of this retrospective study was to describe current real-world treatment and outcomes of pediatric, adolescent, and young adult patients with R/R ALK-positive ALCL. We conducted a retrospective, multi-institutional study identifying 81 patients with R/R ALK-positive ALCL aged ≤21 years at initial diagnosis treated between 2011 and 2022 across 18 institutions. Median time from diagnosis to relapse was 8.9 months (range, 2.6-131.9). Initial reinduction regimens included ALK-inhibitor monotherapy (n = 37, 46%), BV monotherapy (n = 19, 23%), chemotherapy without targeted therapy (n = 12, 15%), chemotherapy with targeted therapy (n = 9, 11%), or vinblastine monotherapy (n = 4, 5%), with 83% of patients achieving a complete response to initial reinduction regimen. Fifty-eight patients received a hematopoietic stem cell transplant (HSCT), 11 autologous and 48 allogeneic, with 1 receiving both. Duration of treatment for patients receiving BV or the ALK-inhibitor crizotinib (CZ) varied widely (BV, 1-11 years; CZ, 2-10 years). Five-year event-free survival was 63% (95% confidence interval [CI], 53-75) and 5-year overall survival was 91% (95% CI, 84-98). This is, to our knowledge, the largest collection of patients with R/R ALK-positive ALCL treated in the era of targeted therapy. Patients achieved excellent responses to ALK-inhibitor or BV monotherapy, but questions remain about duration of therapy and role of HSCT.
Collapse
Affiliation(s)
- Lianna J. Marks
- Division of Pediatric Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Victor Ritter
- Biomedical Informatics Research Division, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Kala Y. Kamdar
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Julie Rivers
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA
| | - Rebecca Gardner
- Department of Oncology and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Matthew J. Ehrhardt
- Department of Oncology and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Kaitlin J. Devine
- Division of Oncology, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA
| | - Charles A. Phillips
- Division of Oncology, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Anne Reilly
- Division of Oncology, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA
| | - Keith August
- Division of Pediatric Hematology, Oncology, and Bone Marrow Transplantation, Children’s Mercy Hospital, Kansas City, MO
| | - Joanna Weinstein
- Department of Hematology, Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Medical Center, New York, NY
| | | | - Christine Moore Smith
- Division of Pediatric Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Chelsee Greer
- Department of Pediatrics, Cook Children’s Medical Center, Fort Worth, TX
| | - Zeinab Afify
- Division of Hematology/Oncology, Department of Pediatrics, The University of Utah, Salt Lake City, UT
| | - Carol H. Lin
- Department of Oncology, Children’s Hospital of Orange County, Orange, CA
| | - Jennifer A. Belsky
- Division of Pediatric Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN
| | - Hilda Ding
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Rady Children’s Hospital, San Diego, CA
| | - David Hoogstra
- Division of Pediatric Hematology/Oncology, Helen DeVos Children’s Hospital, Grand Rapids, MI
| | - Keri Toner
- Division of Oncology, Children’s National Hospital, Washington, DC
| | - Michael P. Link
- Division of Pediatric Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Liora M. Schultz
- Division of Pediatric Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Eric J. Lowe
- Department of Pediatric Hematology/Oncology, Children’s Hospital of the King’s Daughters, Norfolk, VA
| | - Catherine Aftandilian
- Division of Pediatric Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
4
|
Kaloyannidis P, Al-Charfli B, George B, Khalil C, Al-Moghrabi N, Mustafa S, Ibrahim D, Alfar M, Ibrahim F, Odeh B, Daryahya M, Shabo P. AntiCD30-Conjugated Antibody Plus Standard BEAM as Conditioning Regimen for Autologous Hematopoietic Stem Cell Transplantation in Systemic Anaplastic Large Cell Lymphoma. Hematol Rep 2025; 17:3. [PMID: 39846607 PMCID: PMC11755634 DOI: 10.3390/hematolrep17010003] [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: 09/29/2024] [Revised: 12/14/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Background/objectives: The outcome of refractory/relapsed systemic Anaplastic Large Cell Lymphoma (R/R-sALCL), especially for anaplastic lymphoma kinase-1 (ALK-1)-negative disease, remains dismal even after autologous hematopoietic stem cell transplantation (AHSCT). The intensification of both salvage and conditioning regimens, without increasing the toxicity, could improve the outcome of AHSCT in R/R-sALCL. Methods: Based on the successful experience of the incorporation of antiD20 monoclonal antibodies in the treatment of B-Cell Lymphomas, we designed a salvage and conditioning regimen incorporating the antiCD30-conjugated antibody (Brentuximab Vedotin, BV) to standard chemotherapy regimens, and we describe herein the clinical course of a patient with AKL-ve, R/R-sALCL, who received salvage regimen BV + DHAP, followed by AHSCT with preparative regimen consisted of BV plus standard BEAM. Results: The novel regimen was well tolerated, and no severe adverse effects were noticed. The engraftment was prompt and successful. The patient remained in complete metabolic remission for almost 12 months post-transplant. Conclusions: The proposed treatment approach, which combines antiCD30-conjugated antibody with standard salvage and conditioning regimens, demonstrated a completely acceptable toxicity with promising efficacy.
Collapse
Affiliation(s)
- Panayotis Kaloyannidis
- Adult Haematology & Cellular Therapy Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates; (B.A.-C.); (B.G.); (C.K.); (N.A.-M.)
| | - Basmah Al-Charfli
- Adult Haematology & Cellular Therapy Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates; (B.A.-C.); (B.G.); (C.K.); (N.A.-M.)
| | - Biju George
- Adult Haematology & Cellular Therapy Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates; (B.A.-C.); (B.G.); (C.K.); (N.A.-M.)
| | - Charbel Khalil
- Adult Haematology & Cellular Therapy Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates; (B.A.-C.); (B.G.); (C.K.); (N.A.-M.)
| | - Nour Al-Moghrabi
- Adult Haematology & Cellular Therapy Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates; (B.A.-C.); (B.G.); (C.K.); (N.A.-M.)
| | - Samar Mustafa
- Nursing Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates; (S.M.); (M.D.)
| | - Dima Ibrahim
- Infectious Diseases Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates;
| | - Mohammed Alfar
- Pharmacy Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates;
| | - Firuz Ibrahim
- Nuclear Medicine Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates;
| | - Bassam Odeh
- Hematopathology Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates;
| | - Mohammed Daryahya
- Nursing Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates; (S.M.); (M.D.)
| | - Philip Shabo
- Operations Department, Burjeel Medical City, Abu Dhabi 92510, United Arab Emirates;
| |
Collapse
|
5
|
Pichler AS, Amador C, Fujimoto A, Takeuchi K, de Jong D, Iqbal J, Staber PB. Advances in peripheral T cell lymphomas: pathogenesis, genetic landscapes and emerging therapeutic targets. Histopathology 2025; 86:119-133. [PMID: 39679758 DOI: 10.1111/his.15376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Peripheral T cell lymphomas (PTCLs) are a biologically diverse and aggressive group of non-Hodgkin lymphomas that originate from mature T cells, often presenting with complex clinical and morphological features. This review explores the challenges in diagnosing and classifying PTCLs, focusing on the intricate biology of the more common nodal entities. Advances in molecular diagnostics, such as mutational and gene expression profiling, have improved our understanding. However, the rarity and morphological variability of PTCLs continue to complicate the definition of biologically and clinically meaningful entities, as well as the application of current diagnoses in daily practice; these advancements have not yet translated into improved clinical outcomes. Standard therapies fail in most cases and lead to poor prognoses, highlighting the urgent need for improved therapeutic strategies. Precise characterisation of PTCL advances refined classification and supports the development of more targeted and effective treatments. Recent approaches have focused on biology-based risk stratification, either within specific entities or in an entity-agnostic manner. This development aims for improved treatment selection or even personalised treatment based on genetic, epigenetic and functional profiles.
Collapse
Affiliation(s)
- Alexander S Pichler
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Catalina Amador
- Department of Pathology and Laboratory Medicine, University of Miami, Miami, Florida, USA
| | - Ayumi Fujimoto
- Division of Pathology, Cancer Institute, Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Kengo Takeuchi
- Division of Pathology, Cancer Institute, Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Daphne de Jong
- Department of Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Javeed Iqbal
- University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Philipp B Staber
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther 2024; 9:308. [PMID: 39500878 PMCID: PMC11539257 DOI: 10.1038/s41392-024-02004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/19/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024] Open
Abstract
Targeted protein degradation (TPD) represents a revolutionary therapeutic strategy in disease management, providing a stark contrast to traditional therapeutic approaches like small molecule inhibitors that primarily focus on inhibiting protein function. This advanced technology capitalizes on the cell's intrinsic proteolytic systems, including the proteasome and lysosomal pathways, to selectively eliminate disease-causing proteins. TPD not only enhances the efficacy of treatments but also expands the scope of protein degradation applications. Despite its considerable potential, TPD faces challenges related to the properties of the drugs and their rational design. This review thoroughly explores the mechanisms and clinical advancements of TPD, from its initial conceptualization to practical implementation, with a particular focus on proteolysis-targeting chimeras and molecular glues. In addition, the review delves into emerging technologies and methodologies aimed at addressing these challenges and enhancing therapeutic efficacy. We also discuss the significant clinical trials and highlight the promising therapeutic outcomes associated with TPD drugs, illustrating their potential to transform the treatment landscape. Furthermore, the review considers the benefits of combining TPD with other therapies to enhance overall treatment effectiveness and overcome drug resistance. The future directions of TPD applications are also explored, presenting an optimistic perspective on further innovations. By offering a comprehensive overview of the current innovations and the challenges faced, this review assesses the transformative potential of TPD in revolutionizing drug development and disease management, setting the stage for a new era in medical therapy.
Collapse
Affiliation(s)
- Guangcai Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Chang
- School of Pharmaceutical Sciences, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, 450001, China
| | - Weilin Xie
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
7
|
Pereira V, Barthoulot M, Aladjidi N, Contet A, Dalle JH, Dourthe MÉ, Garnier N, Bruno B, Leruste A, Pellier I, Simonin M, Paillard C, Verschuur A, Ducassou S, Lamant L, Brugieres L, Deley MCL, Rigaud C. Outcome of childhood ALK-positive anaplastic large cell lymphoma relapses: Real-life experience of the French Society of Pediatric Oncology (SFCE) cohort of 75 French children. Pediatr Blood Cancer 2024:e31397. [PMID: 39434216 DOI: 10.1002/pbc.31397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVE To describe treatments and outcomes of French children treated for relapsed/refractory anaplastic lymphoma kinase-positive anaplastic large cell lymphoma (ALK+ ALCL). METHODS We conducted the analysis of a series of 75 French children treated for a first relapsed/refractory ALK+ ALCL between 1999 and 2017. RESULTS The median time to first relapse was 8.1 months from initial diagnosis (2.9 after end of treatment), with 12 relapses during frontline treatment or within 1 month of the end of treatment. Treatment of the first relapse varied according to the period of time and risk factors: 48 received multiagent chemotherapy, including 21 and 19 consolidated with allogeneic stem cell transplantation (SCT) and autologous-SCT, respectively. Twenty-one patients received weekly vinblastine, and six received ALK inhibitors (ALKi). Overall, 64/75 patients reached a second complete remission (CR2). Eight out of 11 patients who did not reach CR2 died and the other three were rescued with ALKi, vinblastine, and nivolumab. With a median follow-up of 8.2 years, 60 patients are alive, 43 in CR2, 15 in CR3, two in CR4; and 15 patients died, six from toxicity and nine from disease progression. The 5-year event-free survival and overall survival after first relapse were 51.7% (95% confidence interval [CI]: 39.6%-62.6%) and 80.7% (95% CI: 69.6%-88.1%), respectively. Time to relapse greater than 12 months from initial diagnosis was proven to be a prognostic factor in relapsed/refractory ALK+ ALCL. CONCLUSION In relapsed ALK+ ALCL, high survival rate can be reached with various therapeutic strategies. The main challenge remains to prevent subsequent relapses, and to lower long-term morbidity.
Collapse
Affiliation(s)
- Victor Pereira
- Department of Pediatric Hematology and Oncology, Besançon University Hospital, Besançon, France
- Department of Pediatric and adolescents Oncology, Gustave Roussy cancer Institute, Paris Saclay University, Villejuif, France
| | - Maël Barthoulot
- Department of Methodology and Biostatistics, Oscar Lambret Center, Lille, France
| | - Nathalie Aladjidi
- Department of Pediatric Oncology and Hematology, Bordeaux University Hospital, Bordeaux, France
| | - Audrey Contet
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Nancy, Nancy, France
| | - Jean-Hugues Dalle
- Department of Pediatric Hematology and Immunology, Robert Debré Academic Hospital, GHU AP-HP Nord Université Paris Cité, Paris, France
| | - Marie Émilie Dourthe
- Department of Pediatric Hematology and Immunology, Robert Debré Academic Hospital, GHU AP-HP Nord Université Paris Cité, Paris, France
| | - Nathalie Garnier
- Department of Pediatric Hematology and Oncology, Lyon University Hospital, Lyon, France
| | - Bénédicte Bruno
- Department of Pediatric Hematology, Lille University Hospital, Lille, France
| | - Amaury Leruste
- Department of Pediatric Oncology, Curie Institute, Paris, France
| | - Isabelle Pellier
- Department of Pediatric Oncology and Hematology, Angers University Hospital, Angers, France
| | - Matthieu Simonin
- Department of Pediatric Hematology and Oncology, Trousseau University Hospital, AP-HP, Paris, France
| | - Catherine Paillard
- Department of Pediatric Hematology and Oncology, Strasbourg University Hospital, Strasbourg, France
| | - Arnauld Verschuur
- Department of Pediatric Hematology and Oncology, University Hospital of La Timone, AP-HM, Marseille, France
| | - Stéphane Ducassou
- Department of Pediatric Hematology and Oncology, Bordeaux University Hospital, Bordeaux, France
| | - Laurence Lamant
- Department of Pathology, Cancer University Institute of Toulouse, Toulouse, France
| | - Laurence Brugieres
- Department of Pediatric and adolescents Oncology, Gustave Roussy cancer Institute, Paris Saclay University, Villejuif, France
| | | | - Charlotte Rigaud
- Department of Pediatric and adolescents Oncology, Gustave Roussy cancer Institute, Paris Saclay University, Villejuif, France
| |
Collapse
|
8
|
Adashek JJ, Brodsky M, Levis MJ. Complete morphologic response to gilteritinib in ALK-rearranged acute myeloid leukemia. NPJ Precis Oncol 2024; 8:197. [PMID: 39256524 PMCID: PMC11387604 DOI: 10.1038/s41698-024-00701-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/30/2024] [Indexed: 09/12/2024] Open
Abstract
The cytogenetic abnormality inv(2)(p23q13) in acute myeloid leukemia (AML) results in a fusion of RANBP2 with ALK. This fusion makes ALK constitutively active and acts as a driver for the proliferation of AML cell lines. Gilteritinib, a FLT3 inhibitor approved in AML, also can inhibit ALK among other receptor tyrosine kinases. A 75-year-old-woman with a history of essential thrombocythemia (ET) and a presumed germline DDX41 mutation developed ALK-fusion positive AML and despite standard therapies was transfusion-dependent and globally declining. The patient has been on gilteritinib with an ongoing response of more than one year with near normal blood counts and no evidence of AML. The fact that she was found to harbor a presumed germline DDX41 alteration may account for why she developed, and yet survived, two myeloid neoplasms (ET and AML). Additionally, this demonstrates that gilteritinib is clinically active as an ALK inhibitor, and could be considered for use in any AML patient presenting with an inv(2(p23q13)) translocation. Finally, it is an example of using a disease-agnostic, precision medicine approach to arrive at a beneficial treatment.
Collapse
Affiliation(s)
- Jacob J Adashek
- START Center for Cancer Research - San Antonio, San Antonio, USA.
| | - Max Brodsky
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Mark J Levis
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
9
|
Croci GA, Appio L, Cecchetti C, Tabano S, Alberti-Violetti S, Berti E, Rahal D, Cavallaro F, Onida F, Tomasini D, Todisco E. Primary cutaneous, epidermotropic mycosis fungoides-like presentation: critical appraisal and description of two novel cases, broadening the spectrum of ALK+ T-cell lymphoma. Virchows Arch 2024; 485:417-425. [PMID: 38780617 DOI: 10.1007/s00428-024-03832-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/18/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Leading from a two-case series, including two patients receiving a diagnosis of epidermotropic T-cell lymphoma, featuring a mycosis fungoides (MF)-like clinical pattern and ALK expression and molecular alteration, we performed a critical appraisal of ALK+ primary cutaneous T-cell lymphomas (pcTCL). Considering our patients and the literature, 32 cases were retrieved, 7 of which featured an MF-like clinical picture over a 4-to-20-year period. MF-like cases show distinctive histology, comprising a predominantly epidermotropic infiltration of small-to-large, atypical-to-pleomorphic, with few anaplastic cells, negligible-to-intense CD30-expression, and a CD4+/cytotoxic granule+ phenotype. These features should prompt a search for ALK expression captured by the ALK D5F3 clone. Bona fide ALK+ pcTCL is very rare, and existent data suggest the presence of a broader pattern of disease, including instances mimicking MF and/or primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma. The major challenges in dealing with this subset include prodromal phases, misinterpreted as inflammatory dermatosis or parapsoriasis/early phase MF both clinically and histologically, while recognition of its ALK-driven biology is hampered both by the unusual clinic-pathologic pattern of the disease, which stands apart from the classical (i.e., nodal) picture of ALK+ anaplastic large cell lymphoma and by the low sensitivity of ALK1 clone. Data on its optimal management are far from being conclusive: An MF-like approach is currently chosen, but depending on CD30 and, most notably, ALK expression, a targeted therapy could be envisaged in advanced stages, as clinical response to ALK inhibition was documented in one patient.
Collapse
Affiliation(s)
- Giorgio Alberto Croci
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy.
| | - Lorena Appio
- Division of Hematology, ASST ValleOlona Ospedale di Busto Arsizio, Busto Arsizio, Italy
| | - Caterina Cecchetti
- Division of Hematology, ASST ValleOlona Ospedale di Busto Arsizio, Busto Arsizio, Italy
| | - Silvia Tabano
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Medical Genetics Unit, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Alberti-Violetti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Dermatology Unit, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emilio Berti
- Dermatology Unit, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daoud Rahal
- Department of Pathology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Francesca Cavallaro
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Onida
- Hematology Unit, ASST-Fatebenefratelli-Sacco, University of Milan, Milan, Italy
| | - Dario Tomasini
- Division of Dermatology, ASST ValleOlona Ospedale di Busto Arsizio, Busto Arsizio, Italy
| | - Elisabetta Todisco
- Division of Hematology, ASST ValleOlona Ospedale di Busto Arsizio, Busto Arsizio, Italy
| |
Collapse
|
10
|
Musa S, Amara N, Selawi A, Wang J, Marchini C, Agbarya A, Mahajna J. Overcoming Chemoresistance in Cancer: The Promise of Crizotinib. Cancers (Basel) 2024; 16:2479. [PMID: 39001541 PMCID: PMC11240740 DOI: 10.3390/cancers16132479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Chemoresistance is a major obstacle in cancer treatment, often leading to disease progression and poor outcomes. It arises through various mechanisms such as genetic mutations, drug efflux pumps, enhanced DNA repair, and changes in the tumor microenvironment. These processes allow cancer cells to survive despite chemotherapy, underscoring the need for new strategies to overcome resistance and improve treatment efficacy. Crizotinib, a first-generation multi-target kinase inhibitor, is approved by the FDA for the treatment of ALK-positive or ROS1-positive non-small cell lung cancer (NSCLC), refractory inflammatory (ALK)-positive myofibroblastic tumors (IMTs) and relapsed/refractory ALK-positive anaplastic large cell lymphoma (ALCL). Crizotinib exists in two enantiomeric forms: (R)-crizotinib and its mirror image, (S)-crizotinib. It is assumed that the R-isomer is responsible for the carrying out various processes reviewed here The S-isomer, on the other hand, shows a strong inhibition of MTH1, an enzyme important for DNA repair mechanisms. Studies have shown that crizotinib is an effective multi-kinase inhibitor targeting various kinases such as c-Met, native/T315I Bcr/Abl, and JAK2. Its mechanism of action involves the competitive inhibition of ATP binding and allosteric inhibition, particularly at Bcr/Abl. Crizotinib showed synergistic effects when combined with the poly ADP ribose polymerase inhibitor (PARP), especially in ovarian cancer harboring BRCA gene mutations. In addition, crizotinib targets a critical vulnerability in many p53-mutated cancers. Unlike its wild-type counterpart, the p53 mutant promotes cancer cell survival. Crizotinib can cause the degradation of the p53 mutant, sensitizing these cancer cells to DNA-damaging substances and triggering apoptosis. Interestingly, other reports demonstrated that crizotinib exhibits anti-bacterial activity, targeting Gram-positive bacteria. Also, it is active against drug-resistant strains. In summary, crizotinib exerts anti-tumor effects through several mechanisms, including the inhibition of kinases and the restoration of drug sensitivity. The potential of crizotinib in combination therapies is emphasized, particularly in cancers with a high prevalence of the p53 mutant, such as triple-negative breast cancer (TNBC) and high-grade serous ovarian cancer (HGSOC).
Collapse
Affiliation(s)
- Sanaa Musa
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| | - Noor Amara
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| | - Adan Selawi
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| | - Junbiao Wang
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Cristina Marchini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Abed Agbarya
- Oncology Department, Bnai Zion MC, Haifa 31048, Israel
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 11016, Israel
| |
Collapse
|
11
|
Elhassan E, Girleanu C, Kelly P, Power DG, Sweeney P, Mayer N, Bambury RM. Anaplastic Lymphoma Kinase (ALK)-Rearranged Renal Cell Carcinoma: A Case Report Highlighting Diagnostic Challenges and Therapeutic Opportunities. Cureus 2024; 16:e65621. [PMID: 39205743 PMCID: PMC11350486 DOI: 10.7759/cureus.65621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
A 57-year-old male underwent an open right radical nephrectomy in 2015 for a 3-cm kidney tumor which was classified at the time as a combined tubulocystic and collecting duct carcinoma. One of six nodes was positive for metastatic carcinoma and the patient received adjuvant carboplatin/gemcitabine chemotherapy. In 2020, he developed enlarging retroperitoneal adenopathy and underwent a retroperitoneal lymph node dissection with 11 of 13 nodes in the resected specimen positive for the previously described renal carcinoma, followed by adjuvant radiotherapy. In November 2022, he again underwent surgery for further locoregional recurrence with resection of a right psoas mass lesion and right hemicolectomy. Pathology on this occasion was reclassified as anaplastic lymphoma kinase-rearranged renal cell carcinoma (ALK-RCC). Shortly afterward, a restaging CT revealed multiple liver metastases and evidence of further disease recurrence in the right renal bed. He commenced alectinib with a complete radiological response and has continued on it for 12 months at the time of writing this report. To our knowledge, there are only five prior reports of ALK-RCC treated with targeted ALK inhibitor therapy in the literature. We report this case to highlight the importance of recognizing and diagnosing this rare RCC subtype since it has significant therapeutic implications. Furthermore, to our knowledge, this patient has had the longest follow-up reported to date in the literature so far. A concerted effort by the histopathology and oncology community is needed to gather more data on the incidence and treatment outcomes of these tumors so that progress can be made in optimizing their management. It is important to consider novel and emerging entities from the most recent WHO 2022 classification, many of which are defined by molecular characteristics with associated therapeutic implications.
Collapse
Affiliation(s)
| | | | - Paul Kelly
- Radiation Oncology, Bon Secours Hospital, Cork, IRL
| | - Derek G Power
- Medical Oncology, Cork University Hospital, Cork, IRL
| | | | - Nick Mayer
- Histopathology, Cork University Hospital, Cork, IRL
| | | |
Collapse
|
12
|
Sungwan P, Panaampon J, Kariya R, Kamio S, Nakagawa R, Hirozane T, Ogura Y, Abe M, Hirabayashi K, Fujiwara Y, Kikuta K, Okada S. Establishment and characterization of TK-ALCL1: a novel NPM-ALK-positive anaplastic large-cell lymphoma cell line. Hum Cell 2024; 37:1215-1225. [PMID: 38755432 DOI: 10.1007/s13577-024-01077-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
TK-ALCL1, a novel anaplastic lymphoma kinase (ALK)-positive anaplastic large-cell lymphoma (ALK+ ALCL) cell line, was established from the primary tumor site of a 59-year-old Japanese male patient. The immune profile of TK-ALCL1 corresponds to that seen typically in primary ALCL cells, i.e., positive for ALK, CD30, EMA, and CD4, but negative for CD2, CD3, CD5, CD8a, and EBV-related antigens. The rearrangement of the T cell receptor-gamma locus shows that TK-ALCL1 is clonally derived from T-lineage lymphoid cells. FISH and RT-PCR analysis revealed that TK-ALCL1 has the nucleophosmin (NPM)-ALK fusion transcript, which is typical for ALK+ ALCL cell lines. When TK-ALCL1 was subcutaneously inoculated into 6-week-old BALB/c Rag2-/-/Jak3-/- (BRJ) mice, it formed tumor masses within 4-6 weeks. Morphological, immunohistochemical, and molecular genetic investigations confirmed that the xenograft and the original ALCL tumor were identical. The ALK inhibitors Alectinib and Lorlatinib suppressed proliferation in a dose-dependent manner. Thus, TK-ALCL1 provides a useful in vitro and in vivo model for investigation of the biology of ALK+ ALCL and of novel therapeutic approaches targeting ALK.
Collapse
Affiliation(s)
- Prin Sungwan
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection & Graduate, School of Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuou-ku, Kumamoto, 860-0811, Japan
| | - Jutatip Panaampon
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection & Graduate, School of Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuou-ku, Kumamoto, 860-0811, Japan
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection & Graduate, School of Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuou-ku, Kumamoto, 860-0811, Japan
- Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuou-ku, Kumamoto, 860-8555, Japan
| | - Satoshi Kamio
- Division of Musculoskeletal Oncology and Orthopaedics Surgery, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Rumi Nakagawa
- Division of Musculoskeletal Oncology and Orthopaedics Surgery, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Toru Hirozane
- Division of Musculoskeletal Oncology and Orthopaedics Surgery, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Yukiko Ogura
- Clinical Laboratory Center, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Makoto Abe
- Division of Diagnostic Pathology, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Kaoru Hirabayashi
- Division of Diagnostic Pathology, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuou-ku, Kumamoto, 860-0556, Japan
| | - Kazutaka Kikuta
- Division of Musculoskeletal Oncology and Orthopaedics Surgery, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection & Graduate, School of Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuou-ku, Kumamoto, 860-0811, Japan.
- Institute of Industrial Nanomaterials, Kumamoto University, 2-39-1 Kurokami, Chuou-ku, Kumamoto, 860-8555, Japan.
| |
Collapse
|
13
|
Wasik MA, Kim PM, Nejati R. Diverse and reprogrammable mechanisms of malignant cell transformation in lymphocytes: pathogenetic insights and translational implications. Front Oncol 2024; 14:1383741. [PMID: 38638855 PMCID: PMC11024630 DOI: 10.3389/fonc.2024.1383741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/18/2024] [Indexed: 04/20/2024] Open
Abstract
While normal B- and T-lymphocytes require antigenic ligands to become activated via their B- and T-cell receptors (BCR and TCR, respectively), B- and T-cell lymphomas show the broad spectrum of cell activation mechanisms regarding their dependence on BCR or TCR signaling, including loss of such dependence. These mechanisms are generally better understood and characterized for B-cell than for T-cell lymphomas. While some lymphomas, particularly the indolent, low-grade ones remain antigen-driven, other retain dependence on activation of their antigen receptors seemingly in an antigen-independent manner with activating mutations of the receptors playing a role. A large group of lymphomas, however, displays complete antigen receptor independence, which can develop gradually, in a stepwise manner or abruptly, through involvement of powerful oncogenes. Whereas some of the lymphomas undergo activating mutations of genes encoding proteins involved in signaling cascades downstream of the antigen-receptors, others employ activation mechanisms capable of substituting for these BCR- or TCR-dependent signaling pathways, including reliance on signaling pathways physiologically activated by cytokines. Finally, lymphomas can develop cell-lineage infidelity and in the extreme cases drastically rewire their cell activation mechanisms and engage receptors and signaling pathways physiologically active in hematopoietic stem cells or non-lymphoid cells. Such profound reprograming may involve partial cell dedifferentiation or transdifferentiation towards histocytes, dendritic, or mesodermal cells with various degree of cell maturation along these lineages. In this review, we elaborate on these diverse pathogenic mechanisms underlying cell plasticity and signaling reprogramming as well as discuss the related diagnostic and therapeutic implications and challenges.
Collapse
Affiliation(s)
- Mariusz A. Wasik
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Patricia M. Kim
- Department of Pathology and Laboratory Medicine, Penn State College of Medicine, Hershey, PA, United States
| | - Reza Nejati
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, United States
| |
Collapse
|
14
|
Liu W, Wu J, Ming X, Zhang Q, Zhou D, Zheng R, Zhou M, Shang Z, Chen L, Zhu X, Xiao Y. Case report: The utilization of crizotinib and brentuximab vedotin as a bridge to autologous stem cell transplantation and followed by CD30-directed CAR-T cell therapy in relapsed/refractory ALK+ ALCL. Front Immunol 2024; 15:1346001. [PMID: 38375471 PMCID: PMC10875076 DOI: 10.3389/fimmu.2024.1346001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
Background Anaplastic lymphoma kinase-positive anaplastic large cell lymphoma (ALK+ ALCL) is a rare, mature T-cell non-Hodgkin lymphoma. The prognosis of patients with relapsed or refractory ALCL following first-line chemotherapy is extremely poor. NCCN guidelines recommend intensified chemotherapy with or without ASCT consolidation for r/r ALCL, however, this is not an effective treatment for all ALK+ALCL. Case report Herein, we report a patient with relapsed/refractory ALK+ ALCL who received crizotinib and brentuximab vedotin as bridging therapy, followed by autologous stem cell transplantation and sequential anti-CD30 CAR T cell therapy. Conclusion The patient achieved complete remission and long-term disease-free survival of months and continues to be followed up. The combination therapy model in this case may provide guidance for the management of relapsed/refractory ALK+ ALCL, and further prospective trials are needed to confirm its effectiveness.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|