239 results on '"Irina Maric"'
Search Results
2. Publisher Correction: Oncogenic RAS commandeers amino acid sensing machinery to aberrantly activate mTORC1 in multiple myeloma
- Author
-
Yandan Yang, Arnold Bolomsky, Thomas Oellerich, Ping Chen, Michele Ceribelli, Björn Häupl, George W. Wright, James D. Phelan, Da Wei Huang, James W. Lord, Callie K. Van Winkle, Xin Yu, Jan Wisniewski, James Q. Wang, Frances A. Tosto, Erin Beck, Kelli Wilson, Crystal McKnight, Jameson Travers, Carleen Klumpp-Thomas, Grace A. Smith, Stefania Pittaluga, Irina Maric, Dickran Kazandjian, Craig J. Thomas, and Ryan M. Young
- Subjects
Science - Published
- 2022
- Full Text
- View/download PDF
3. Myelodysplasia in the setting of paroxysmal nocturnal hemoglobinuria: Interpretation of blast percentage in a marrow with erythroid hyperplasia
- Author
-
Emma M. Groarke, Bhavisha A. Patel, Alina Dulau‐Florea, Irina Maric, Neal S. Young, and Katherine R. Calvo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
4. Corrigendum: Paradoxical CD4 Lymphopenia in Autoimmune Lymphoproliferative Syndrome (ALPS)
- Author
-
Andrea Lisco, Chun-Shu Wong, Susan Price, Peiying Ye, Julie Niemela, Megan Anderson, Elizabeth Richards, Maura Manion, Harry Mystakelis, Morgan Similuk, Bernice Lo, Jennifer Stoddard, Sergio Rosenzweig, Christophe Vanpouille, Adam Rupert, Irina Maric, Ainhoa Perez-Diez, David Parenti, Peter D. Burbelo, V. Koneti Rao, and Irini Sereti
- Subjects
CD4 lymphopenia ,follicular T helper cells ,ALPS-FAS ,autoimmune cytopenia ,apoptosis ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2019
- Full Text
- View/download PDF
5. Paradoxical CD4 Lymphopenia in Autoimmune Lymphoproliferative Syndrome (ALPS)
- Author
-
Andrea Lisco, Chun-Shu Wong, Susan Price, Peiying Ye, Julie Niemela, Megan Anderson, Elizabeth Richards, Maura Manion, Harry Mystakelis, Morgan Similuk, Bernice Lo, Jennifer Stoddard, Sergio Rosenzweig, Christophe Vanpouille, Adam Rupert, Irina Maric, Ainhoa Perez-Diez, David Parenti, Peter D. Burbelo, V. Koneti Rao, and Irini Sereti
- Subjects
CD4 lymphopenia ,follicular T helper cells ,ALPS-FAS ,autoimmune cytopenia ,apoptosis ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is caused by germline or somatic loss of function FAS mutations resulting in impaired apoptosis and consequent expansion of T-lymphocytes causing organomegaly and autoimmune anemia, neutropenia and thrombocytopenia. Herein, we report on a case of disseminated varicella zoster infection after post-partum vaccination in a patient found to have CD4 lymphopenia and eventually diagnosed with ALPS caused by a novel germline missense mutation in FAS death-domain. A subsequent retrospective analysis of 169 patients of the NIH ALPS-FAS cohort, revealed that CD4-T-cells lymphopenia (< 300 cells/μl) may occur in 5% of ALPS-FAS patients irrespectively of the underlying genetic defect, organomegaly or immunosuppressive treatment. Although immunophenotyping did not show depletion of specific CD4-T-cells subpopulations, CD4-lymphopenic ALPS-FAS subjects had an expansion of a subset of circulating T-follicular-helper (cTfh) cells, associated with autoantibody production (CCR7lowPD-1high). Furthermore, autoantibodies binding on CD4-T-cells were detected in 50% of the CD4-lymphopenic ALPS-FAS patients and caused cytotoxicity in a natural killer (NK)-mediated antibody-dependent-cellular cytotoxicity assay. Such autoantibodies can therefore be associated with CD4-T-cell death, impaired activation induced proliferation or impaired trafficking. The expansion of autoreactive T-cells in ALPS-FAS is known to be associated with autoimmune clinical manifestations, however our study reveals that ALPS-FAS can also be associated with a paradoxical depletion of CD4-T-cells due to the presence of autoantibodies on the surface of CD4-T-cells which can in turn result in increased susceptibility to opportunistic infections. These novel findings have implications for the diagnosis, clinical monitoring, and management of patients with ALPS-FAS.
- Published
- 2019
- Full Text
- View/download PDF
6. Bone marrow findings in autoimmune lymphoproliferative syndrome with germline FAS mutation
- Author
-
Yi Xie, Stefania Pittaluga, Susan Price, Mark Raffeld, Jamie Hahn, Elaine S. Jaffe, V. Koneti Rao, and Irina Maric
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Autoimmune lymphoproliferative syndrome is a rare genetic disorder characterized by defective FAS-mediated apoptosis, autoimmune disease, accumulation of mature T-cell receptor alpha/beta positive, CD4 and CD8 double-negative T cells and increased risk of lymphoma. Despite frequent hematologic abnormalities, literature is scarce regarding the bone marrow pathology in autoimmune lymphoproliferative syndrome. We retrospectively reviewed 3l bone marrow biopsies from a cohort of 240 patients with germline FAS mutations. All biopsies were performed for the evaluation of cytopenias or to rule out lymphoma. Clinical information was collected and morphological, immunohistochemical, flow cytometric and molecular studies were performed. Bone marrow lymphocytosis was the predominant feature, present in 74% (23/31) of biopsies. The lymphoid cells showed several different patterns of infiltration, most often forming aggregates comprising T cells in 15 cases, B cells in one and a mixture of T and B cells in the other seven cases. Double-negative T cells were detected by immunohistochemistry in the minority of cases (10/31; 32%); significantly, all but one of these cases had prominent double-negative T-lymphoid aggregates, which in four cases diffusely replaced the marrow space. One case showed features of Rosai-Dorfman disease, containing scattered S-100+ cells with emperipolesis and double-negative T cells. No clonal B or T cells were detected by polymerase chain reaction in any evaluated cases. Classical Hodgkin lymphoma was identified in three cases. Our results demonstrate that infiltrates of T cells, or rarely B cells, can be extensive in patients with autoimmune lymphoproliferative syndrome, mimicking lymphoma. A multi-modality approach, integrating clinical, histological, immunohistochemical as well as other ancillary tests, can help avoid this diagnostic pitfall. This study is registered at Clinicaltrials.gov ID # NCT00001350
- Published
- 2017
- Full Text
- View/download PDF
7. Paradoxical resistance of multiple myeloma to proteasome inhibitors by decreased levels of 19S proteasomal subunits
- Author
-
Diego Acosta-Alvear, Min Y Cho, Thomas Wild, Tonia J Buchholz, Alana G Lerner, Olga Simakova, Jamie Hahn, Neha Korde, Ola Landgren, Irina Maric, Chunaram Choudhary, Peter Walter, Jonathan S Weissman, and Martin Kampmann
- Subjects
proteasome ,cancer ,proteostasis ,carfilzomib ,myeloma ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Hallmarks of cancer, including rapid growth and aneuploidy, can result in non-oncogene addiction to the proteostasis network that can be exploited clinically. The defining example is the exquisite sensitivity of multiple myeloma (MM) to 20S proteasome inhibitors, such as carfilzomib. However, MM patients invariably acquire resistance to these drugs. Using a next-generation shRNA platform, we found that proteostasis factors, including chaperones and stress-response regulators, controlled the response to carfilzomib. Paradoxically, 19S proteasome regulator knockdown induced resistance to carfilzomib in MM and non-MM cells. 19S subunit knockdown did not affect the activity of the 20S subunits targeted by carfilzomib nor their inhibition by the drug, suggesting an alternative mechanism, such as the selective accumulation of protective factors. In MM patients, lower 19S levels predicted a diminished response to carfilzomib-based therapies. Together, our findings suggest that an understanding of network rewiring can inform development of new combination therapies to overcome drug resistance.
- Published
- 2015
- Full Text
- View/download PDF
8. Episodic angioedema with eosinophilia (Gleich syndrome) is a multilineage cell cycling disorder
- Author
-
Paneez Khoury, Jacqueline Herold, Alexandra Alpaugh, Ellen Dinerman, Nicole Holland-Thomas, Jennifer Stoddard, Shakuntala Gurprasad, Irina Maric, Olga Simakova, Lawrence B. Schwartz, Juelia Fong, Chyi-Chia Richard Lee, Liqiang Xi, Zengfeng Wang, Mark Raffeld, and Amy D. Klion
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Episodic angioedema with eosinophilia (Gleich syndrome) is a rare disorder characterized by episodes of angioedema and eosinophilia that occur at monthly intervals and resolve spontaneously without therapy. Despite the striking periodicity of this disorder, its similarity to other cyclic hematopoietic disorders with multilineage involvement has not been assessed. To characterize the involvement of cell lineages in the etiology and pathogenesis of episodic angioedema with eosinophilia, four subjects were evaluated by blood counts and other analyses over the course of 1–2 months. Surface marker expression was assessed on T cells by flow cytometry and clonality by polymerase chain reaction. Intracellular cytokine evaluation, bone marrow and skin biopsies were performed during different parts of the cycle. Cycling of multiple cell lineages, including neutrophils, lymphocytes and eosinophils, was observed in the four subjects with the disorder with a periodicity of 25–35 days. An aberrant CD3−CD4+ T-cell population was detected in all four subjects, and T-cell receptor rearrangement studies showed a clonal pattern in three subjects. A peak of type II cytokines was detected in the serum of subjects prior to the onset of symptoms and eosinophil cycling and corresponded to ex-vivo type II cytokines detected intracellularly in CD3+CD4+CD154+ T cells. Although the etiology of episodic angioedema with eosinophilia is not yet known, multiple lineages, including lymphocytes, neutrophils and mast cells, are involved and may be related to disease pathogenesis. Whether these cells act directly or promote eosinophilia and eosinophil activation remains to be elucidated. All subjects gave informed consent and were evaluated under an Institutional Review Board-approved protocol (NCT00001406).
- Published
- 2015
- Full Text
- View/download PDF
9. A phase II trial of pan-KIR2D blockade with IPH2101 in smoldering multiple myeloma
- Author
-
Neha Korde, Mattias Carlsten, Min-Jung Lee, Alex Minter, Esther Tan, Mary Kwok, Elisabet Manasanch, Manisha Bhutani, Nishant Tageja, Mark Roschewski, Adriana Zingone, Rene Costello, Marcia Mulquin, Diamond Zuchlinski, Irina Maric, Katherine R. Calvo, Raul Braylan, Prashant Tembhare, Constance Yuan, Maryalice Stetler-Stevenson, Jane Trepel, Richard Childs, and Ola Landgren
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2014
- Full Text
- View/download PDF
10. MicroRNA profiling of follicular lymphoma identifies microRNAs related to cell proliferation and tumor response
- Author
-
Weixin Wang, Meghan Corrigan-Cummins, Justin Hudson, Irina Maric, Olga Simakova, Sattva S. Neelapu, Larry W. Kwak, John E. Janik, Barry Gause, Elaine S. Jaffe, and Katherine R. Calvo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background MicroRNAs can play an important role in tumorigenesis through post-transcriptional regulation of gene expression, and are not well characterized in follicular lymphoma.Design and Methods MicroRNA profiles of enriched follicular lymphoma tumor cells from 16 patients were generated by assaying 851 human microRNAs. Tandem gene expression profiles were obtained for predicting microRNA targets.Results The expression of 133 microRNAs was significantly different (> 2-fold; P
- Published
- 2012
- Full Text
- View/download PDF
11. Myelodysplasia in autosomal dominant and sporadic monocytopenia immunodeficiency syndrome: diagnostic features and clinical implications
- Author
-
Katherine R. Calvo, Donald C. Vinh, Irina Maric, Weixin Wang, Pierre Noel, Maryalice Stetler-Stevenson, Diane C. Arthur, Mark Raffeld, Amalia Dutra, Evgenia Pak, Kyungjae Myung, Amy P. Hsu, Dennis D. Hickstein, Stefania Pittaluga, and Steven M. Holland
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
A novel, genetic immunodeficiency syndrome has been recently described, herein termed “MonoMAC”. It is characterized by severe circulating monocytopenia, NK- and B-lymphocytopenia, severe infections with M. avium complex (MAC), and risk of progression to myelodysplasia/acute myelogenous leukemia. Detailed bone marrow analyses performed on 18 patients further define this disorder. The majority of patients had hypocellular marrows with reticulin fibrosis and multilineage dysplasia affecting the myeloid (72%), erythroid (83%) and megakaryocytic (100%) lineages. Cytogenetic abnormalities were present in 10 of 17 (59%). Despite B-lymphocytopenia, plasma cells were present but were abnormal (e.g. CD56+) in nearly half of cases. Increased T-cell large granular lymphocyte populations were present in 28% of patients. Chromosomal breakage studies, cell cycle checkpoint functions, and sequencing of TERT and K-RAS genes revealed no abnormalities. MonoMAC appears to be a unique, inherited syndrome of bone marrow failure. We describe distinctive bone marrow features to help in its recognition and diagnosis. (Clinicaltrials.gov identifiers: NCT00018044, NCT00923364, NCT01212055)
- Published
- 2011
- Full Text
- View/download PDF
12. Clonal analysis of NRAS activating mutations in KIT-D816V systemic mastocytosis
- Author
-
Todd M. Wilson, Irina Maric, Olga Simakova, Yun Bai, Eunice Ching Chan, Nicolas Olivares, Melody Carter, Dragan Maric, Jamie Robyn, and Dean D. Metcalfe
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Cooperating genetic events are likely to contribute to the phenotypic diversity of KIT-D816V systemic mastocytosis. In this study, 44 patients with KIT-D816V systemic mastocytosis were evaluated for coexisting NRAS, KRAS, HRAS or MRAS mutations. Activating NRAS mutations were identified in 2 of 8 patients with advanced disease. NRAS mutations were not found in patients with indolent systemic mastocytosis. To better understand the clonal evolution of mastocytosis, we evaluated the cell compartments impacted by the NRAS and KIT mutations. Clonal mast cells harbored both mutations. KIT-D816V was not detected in bone marrow CD34+ progenitors, whereas the NRAS mutation was present. These findings suggest that NRAS mutations may have the potential to precede KIT-D816V in clonal development. Unlike other mature lineages, mast cell survival is dependent on KIT and the presence of these two activating mutations may have a greater impact on the expansion of this cell compartment and in resultant disease severity. (Clinicaltrials.gov identifier: NCT00044122, NCT00001756)
- Published
- 2011
- Full Text
- View/download PDF
13. Activity of imatinib in systemic mastocytosis with chronic basophilic leukemia and a PRKG2-PDGFRB fusion
- Author
-
Idoya Lahortiga, Cem Akin, Jan Cools, Todd M. Wilson, Nicole Mentens, Diane C. Arthur, Irina Maric, Pierre Noel, Can Kocabas, Peter Marynen, Lawrence S. Lessin, Iwona Wlodarska, Jamie Robyn, and Dean D. Metcalfe
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background Translocations involving region 5q31-32 (PDGFRB) have been reported in a variety of myeloproliferative diseases and are often associated with significant peripheral eosinophilia. We report an unusual case of a patient presenting with peripheral basophilia and systemic mastocytosis in whom cytogenetic analysis revealed a t(4;5)(q21.1;q31.3).Design and Methods We used molecular analyses to determine the role of PDGFRB in this case. The patient was treated with imatinib.Results Fluorescence in situ hybridization (FISH) documented a breakpoint in PDGFRB. In agreement with this, the patient responded very well to imatinib with resolution of clinical symptoms, basophilia, and mast cell disease. Molecular analyses revealed that PDGFRB, encoding an imatinib-sensitive tyrosine kinase, was fused to PRKG2. The fusion gene incorporates the first two exons of PRKG2 fused to the truncated exon 12 of PDGFRB, resulting in the disruption of its juxtamembrane domain. Functional studies confirmed that the activity and transforming properties of PRKG2-PDGFRβ were dependent on the disruption of the auto-inhibitory juxtamembrane domain.Conclusions Our results identify a second case of the PRKG2-PDGFRB fusion and confirm the unusual PDGFRB breakpoint associated with this fusion. This work also illustrates the use of imatinib for the treatment of specific cases of systemic mastocytosis.
- Published
- 2008
- Full Text
- View/download PDF
14. von Hippel-Lindau disease-associated hemangioblastomas are derived from embryologic multipotent cells.
- Author
-
Deric M Park, Zhengping Zhuang, Ling Chen, Nicholas Szerlip, Irina Maric, Jie Li, Taesung Sohn, Stephanie H Kim, Irina A Lubensky, Alexander O Vortmeyer, Griffin P Rodgers, Edward H Oldfield, and Russell R Lonser
- Subjects
Medicine - Abstract
BACKGROUND: To determine the origin of the neoplastic cell in central nervous system (CNS) hemangioblastomas in von Hippel-Lindau disease (VHL) and its role in tumor formation and distribution, we characterized and differentiated neoplastic cells from hemangioblastomas removed from VHL patients. METHODS AND FINDINGS: A total of 31 CNS hemangioblastomas from 25 VHL patients were resected and analyzed. Tumor cells from the hemangioblastomas were characterized, grown, and differentiated into multiple lineages. Resected hemangioblastomas were located in the cerebellum (11 tumors), brainstem (five tumors), and spinal cord (15 tumors). Consistent with an embryologically derived hemangioblast, the neoplastic cells demonstrated coexpression of the mesodermal markers brachyury, Flk-1 (vascular endothelial growth factor-2), and stem cell leukemia (Scl). The neoplastic cells also expressed hematopoietic stem cell antigens and receptors including CD133, CD34, c-kit, Scl, erythropoietin, and erythropoietin receptor. Under specific microenvironments, neoplastic cells (hemangioblasts) were expanded and differentiated into erythrocytic, granulocytic, and endothelial progenitors. Deletion of the wild-type VHL allele in the hematopoietic and endothelial progeny confirmed their neoplastic origin. CONCLUSIONS: The neoplastic cell of origin for CNS hemangioblastomas in VHL patients is the mesoderm-derived, embryologically arrested hemangioblast. The hematopoietic and endothelial differentiation potential of these cells can be reactivated under suitable conditions. These findings may also explain the unique tissue distribution of tumor involvement.
- Published
- 2007
- Full Text
- View/download PDF
15. Genetically defined individual reference ranges for tryptase limit unnecessary procedures and unmask myeloid neoplasms
- Author
-
Jack Chovanec, Ilker Tunc, Jason Hughes, Joseph Halstead, Allyson Mateja, Yihui Liu, Michael P. O’Connell, Jiwon Kim, Young Hwan Park, Qinlu Wang, Quang Le, Mehdi Pirooznia, Neil N. Trivedi, Yun Bai, Yuzhi Yin, Amy P. Hsu, Joshua McElwee, Sheryce Lassiter, Celeste Nelson, Judy Bandoh, Thomas DiMaggio, Julij Šelb, Matija Rijavec, Melody C. Carter, Hirsh D. Komarow, Vito Sabato, Joshua Steinberg, Kurt M. Hafer, Elizabeth Feuille, Christopher S. Hourigan, Justin Lack, Paneez Khoury, Irina Maric, Roberta Zanotti, Patrizia Bonadonna, Lawrence B. Schwartz, Joshua D. Milner, Sarah C. Glover, Didier G. Ebo, Peter Korošec, George H. Caughey, Erica H. Brittain, Ben Busby, Dean D. Metcalfe, and Jonathan J. Lyons
- Subjects
Hematology - Abstract
Serum tryptase is a biomarker used to aid in the identification of certain myeloid neoplasms, most notably systemic mastocytosis, where basal serum tryptase (BST) levels >20 ng/mL are a minor criterion for diagnosis. Although clonal myeloid neoplasms are rare, the common cause for elevated BST levels is the genetic trait hereditary α-tryptasemia (HαT) caused by increased germline TPSAB1 copy number. To date, the precise structural variation and mechanism(s) underlying elevated BST in HαT and the general clinical utility of tryptase genotyping, remain undefined. Through cloning, long-read sequencing, and assembling of the human tryptase locus from an individual with HαT, and validating our findings in vitro and in silico, we demonstrate that BST elevations arise from overexpression of replicated TPSAB1 loci encoding canonical α-tryptase protein owing to coinheritance of a linked overactive promoter element. Modeling BST levels based on TPSAB1 replication number, we generate new individualized clinical reference values for the upper limit of normal. Using this personalized laboratory medicine approach, we demonstrate the clinical utility of tryptase genotyping, finding that in the absence of HαT, BST levels >11.4 ng/mL frequently identify indolent clonal mast cell disease. Moreover, substantial BST elevations (eg, >100 ng/mL), which would ordinarily prompt bone marrow biopsy, can result from TPSAB1 replications alone and thus be within normal limits for certain individuals with HαT.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table 1 from Interactions between Ibrutinib and Anti-CD20 Antibodies: Competing Effects on the Outcome of Combination Therapy
- Author
-
Adrian Wiestner, Sarah E.M. Herman, Mohammed Z.H. Farooqui, Susan Soto, Janet Valdez, Constance Yuan, Katherine R. Calvo, Gerald E. Marti, Maryalice Stetler-Stevenson, Dalia Salem, Irina Maric, Sabrina Martyr, Yuh Shan Lee, Carsten U. Niemann, and Martin Skarzynski
- Abstract
Supplementary Table 1. Characteristics of all patients studied.
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Interactions between Ibrutinib and Anti-CD20 Antibodies: Competing Effects on the Outcome of Combination Therapy
- Author
-
Adrian Wiestner, Sarah E.M. Herman, Mohammed Z.H. Farooqui, Susan Soto, Janet Valdez, Constance Yuan, Katherine R. Calvo, Gerald E. Marti, Maryalice Stetler-Stevenson, Dalia Salem, Irina Maric, Sabrina Martyr, Yuh Shan Lee, Carsten U. Niemann, and Martin Skarzynski
- Abstract
Purpose: Clinical trials of ibrutinib combined with anti-CD20 monoclonal antibodies (mAb) for chronic lymphocytic leukemia (CLL) report encouraging results. Paradoxically, in preclinical studies, in vitro ibrutinib was reported to decrease CD20 expression and inhibit cellular effector mechanisms. We therefore set out to investigate effects of in vivo ibrutinib treatment that could explain this paradox.Experimental Design: Patients received single-agent ibrutinib (420 mg daily) on an investigator-initiated phase II trial. Serial blood samples were collected pretreatment and during treatment for ex vivo functional assays to examine the effects on CLL cell susceptibility to anti-CD20 mAbs.Results: We demonstrate that CD20 expression on ibrutinib was rapidly and persistently downregulated (median reduction 74%, day 28, P < 0.001) compared with baseline. Concomitantly, CD20 mRNA was decreased concurrent with reduced NF-κB signaling. An NF-κB binding site in the promoter of MS4A1 (encoding CD20) and downregulation of CD20 by NF-κB inhibitors support a direct transcriptional effect. Ex vivo, tumor cells from patients on ibrutinib were less susceptible to anti-CD20 mAb-mediated complement-dependent cytotoxicity than pretreatment cells (median reduction 75%, P < 0.001); however, opsonization by the complement protein C3d, which targets cells for phagocytosis, was relatively maintained. Expression of decay-accelerating factor (CD55) decreased on ibrutinib, providing a likely mechanism for the preserved C3d opsonization. In addition, ibrutinib significantly inhibited trogocytosis, a major contributor to antigen loss and tumor escape during mAb therapy.Conclusions: Our data indicate that ibrutinib promotes both positive and negative interactions with anti-CD20 mAbs, suggesting that successfully harnessing maximal antitumor effects of such combinations requires further investigation. Clin Cancer Res; 22(1); 86–95. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figures from Interactions between Ibrutinib and Anti-CD20 Antibodies: Competing Effects on the Outcome of Combination Therapy
- Author
-
Adrian Wiestner, Sarah E.M. Herman, Mohammed Z.H. Farooqui, Susan Soto, Janet Valdez, Constance Yuan, Katherine R. Calvo, Gerald E. Marti, Maryalice Stetler-Stevenson, Dalia Salem, Irina Maric, Sabrina Martyr, Yuh Shan Lee, Carsten U. Niemann, and Martin Skarzynski
- Abstract
Supplementary Figure 1. Uniform CD20 loss from the surface of primary CLL cells after in vivo exposure to ibrutinib. Supplementary Figure 2. Loss of CD20 on ibrutinib and basal CD20 expression levels do not correlate with high risk prognostic factors. Supplementary Figure 3. Selective NF-κB inhibitor diminishes CD20 cell surface expression. Supplementary Figure 4. Complement regulatory proteins membrane cofactor protein and protectin are not consistently inhibited by ibrutinib.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary figures S1-S5 from Disruption of in vivo Chronic Lymphocytic Leukemia Tumor–Microenvironment Interactions by Ibrutinib – Findings from an Investigator-Initiated Phase II Study
- Author
-
Adrian Wiestner, Mohammed Z.H. Farooqui, Gerald E. Marti, Clare Sun, Jade Jones, Deanna H. Wong, Yuh Shan Lee, Janet Valdez, Raul C. Braylan, Katherine R. Calvo, Constance M. Yuan, Maryalice Stetler-Stevenson, Sabrina Martyr, Betty Y. Chang, Angelique Biancotto, Julio Gomez-Rodriguez, Irina Maric, Sarah E.M. Herman, and Carsten U. Niemann
- Abstract
Supplementary Figure S1. Ibrutinib alters the absolute expression of inflammatory cytokines and chemokines in serum from patients with CLL. Supplementary Figure S2. Ibrutinib treatment reduces tumor burden. Supplementary Figure S3. Both CD4+ and CD8+ T cell subsets are decreased on ibrutinib. Supplementary Figure S4. Grading of macrophage interaction with CLL cells in bone marrow specimens. Supplementary Figure S5. Ibrutinib alters the absolute expression of chemokines in bone marrow supernatant from patients with CLL.
- Published
- 2023
- Full Text
- View/download PDF
20. Data from Disruption of in vivo Chronic Lymphocytic Leukemia Tumor–Microenvironment Interactions by Ibrutinib – Findings from an Investigator-Initiated Phase II Study
- Author
-
Adrian Wiestner, Mohammed Z.H. Farooqui, Gerald E. Marti, Clare Sun, Jade Jones, Deanna H. Wong, Yuh Shan Lee, Janet Valdez, Raul C. Braylan, Katherine R. Calvo, Constance M. Yuan, Maryalice Stetler-Stevenson, Sabrina Martyr, Betty Y. Chang, Angelique Biancotto, Julio Gomez-Rodriguez, Irina Maric, Sarah E.M. Herman, and Carsten U. Niemann
- Abstract
Purpose: Chronic lymphocytic leukemia (CLL) cells depend on microenvironmental interactions for proliferation and survival that are at least partially mediated through B-cell receptor (BCR) signaling. Ibrutinib, a Bruton tyrosine kinase inhibitor, disrupts BCR signaling and leads to the egress of tumor cells from the microenvironment. Although the on-target effects on CLL cells are well defined, the impact on the microenvironment is less well studied. We therefore sought to characterize the in vivo effects of ibrutinib on the tumor microenvironment.Experimental Design: Patients received single-agent ibrutinib on an investigator-initiated phase II trial. Serial blood and tissue samples were collected pretreatment and during treatment. Changes in cytokine levels, cellular subsets, and microenvironmental interactions were assessed.Results: Serum levels of key chemokines and inflammatory cytokines decreased significantly in patients on ibrutinib. Furthermore, ibrutinib treatment decreased circulating tumor cells and overall T-cell numbers. Most notably, a reduced frequency of the Th17 subset of CD4+ T cells was observed concurrent with reduced expression of activation markers and PD-1 on T cells. Consistent with direct inhibition of T cells, ibrutinib inhibited Th17 differentiation of murine CD4+ T cells in vitro. Finally, in the bone marrow microenvironment, we found that ibrutinib disaggregated the interactions of macrophages and CLL cells, inhibited secretion of CXCL13, and decreased the chemoattraction of CLL cells.Conclusions: In conjunction with inhibition of BCR signaling, these changes in the tumor microenvironment likely contribute to the antitumor activity of ibrutinib and may impact the efficacy of immunotherapeutic strategies in patients with CLL. Clin Cancer Res; 22(7); 1572–82. ©2015 AACR.See related commentary by Bachireddy and Wu, p. 1547
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary tables and figure legends from Disruption of in vivo Chronic Lymphocytic Leukemia Tumor–Microenvironment Interactions by Ibrutinib – Findings from an Investigator-Initiated Phase II Study
- Author
-
Adrian Wiestner, Mohammed Z.H. Farooqui, Gerald E. Marti, Clare Sun, Jade Jones, Deanna H. Wong, Yuh Shan Lee, Janet Valdez, Raul C. Braylan, Katherine R. Calvo, Constance M. Yuan, Maryalice Stetler-Stevenson, Sabrina Martyr, Betty Y. Chang, Angelique Biancotto, Julio Gomez-Rodriguez, Irina Maric, Sarah E.M. Herman, and Carsten U. Niemann
- Abstract
Supplementary Table S1: Evaluation of serum cytokine levels on ibrutinib Supplementary Table S2: Evaluation of chemoattractant levels in bone marrow supernatant on ibrutinib supplementary figure legends for supplementary figures S1-S5
- Published
- 2023
- Full Text
- View/download PDF
22. Conflict of Interest Form from Disruption of in vivo Chronic Lymphocytic Leukemia Tumor–Microenvironment Interactions by Ibrutinib – Findings from an Investigator-Initiated Phase II Study
- Author
-
Adrian Wiestner, Mohammed Z.H. Farooqui, Gerald E. Marti, Clare Sun, Jade Jones, Deanna H. Wong, Yuh Shan Lee, Janet Valdez, Raul C. Braylan, Katherine R. Calvo, Constance M. Yuan, Maryalice Stetler-Stevenson, Sabrina Martyr, Betty Y. Chang, Angelique Biancotto, Julio Gomez-Rodriguez, Irina Maric, Sarah E.M. Herman, and Carsten U. Niemann
- Abstract
Conflict of Interest Form from Disruption of in vivo Chronic Lymphocytic Leukemia Tumor–Microenvironment Interactions by Ibrutinib – Findings from an Investigator-Initiated Phase II Study
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figures Legends from Interactions between Ibrutinib and Anti-CD20 Antibodies: Competing Effects on the Outcome of Combination Therapy
- Author
-
Adrian Wiestner, Sarah E.M. Herman, Mohammed Z.H. Farooqui, Susan Soto, Janet Valdez, Constance Yuan, Katherine R. Calvo, Gerald E. Marti, Maryalice Stetler-Stevenson, Dalia Salem, Irina Maric, Sabrina Martyr, Yuh Shan Lee, Carsten U. Niemann, and Martin Skarzynski
- Abstract
Supplementary Figures Legends from Interactions between Ibrutinib and Anti-CD20 Antibodies: Competing Effects on the Outcome of Combination Therapy
- Published
- 2023
- Full Text
- View/download PDF
24. Challenges in Applying Diagnostic Criteria for Systemic Mastocytosis
- Author
-
Nathan A Boggs, Xiaoping Sun, Jonathan J Lyons, Jeremy C McMurray, David M Rose, Eric M Pryor, Dean D. Metcalfe, and Irina Maric
- Subjects
Hematology - Published
- 2023
- Full Text
- View/download PDF
25. Genetically determining individualized clinical reference ranges for the biomarker tryptase can limit unnecessary procedures and unmask myeloid neoplasms
- Author
-
Jack Chovanec, Ilker Tunc, Jason Hughes, Joseph Halstead, Allyson Mateja, Yihui Liu, Michael P. O’Connell, Jiwon Kim, Young Hwan Park, Qinlu Wang, Quang Le, Mehdi Pirooznia, Neil N. Trivedi, Yun Bai, Yuzhi Yin, Amy P. Hsu, Josh McElwee, Sheryce Lassiter, Celeste Nelson, Judy Bandoh, Thomas DiMaggio, Julij Šelb, Matija Rijavec, Melody C. Carter, Hirsh D. Komarow, Vito Sabato, Joshua Steinberg, Kurt M. Hafer, Elizabeth Feuille, Christopher S. Hourigan, Justin Lack, Paneez Khoury, Irina Maric, Roberta Zanotti, Patrizia Bonadonna, Lawrence B. Schwartz, Joshua D. Milner, Sarah C. Glover, Didier G. Ebo, Peter Korošec, George H. Caughey, Erica H. Brittain., Ben Busby, Dean D. Metcalfe, and Jonathan J. Lyons
- Subjects
Human medicine - Abstract
Serum tryptase is a biomarker used to aid in the identification of certain myeloid neoplasms, most notably systemic mastocytosis, where baseline (BST) levels >20 ng/mL are a minor criterion for diagnosis. Whereas clonal myeloid neoplasms are rare, the common cause for elevated BST is the genetic trait hereditary alpha-tryptasemia (HαT) caused by increased germline TPSAB1 copy number. To date, the precise structural variation and mechanism(s) underlying elevated BST in HαT and the general clinical utility of tryptase genotyping, remain undefined. Through cloning, long-read sequencing, and assembling of the human tryptase locus from an individual with HαT, and validating our findings in vitro and in silico, we demonstrate that BST elevations arise from over-expression of replicated TPSAB1 loci encoding wild-type α-tryptase due to co-inheritance of a linked over-active promoter element. Modeling BST levels based upon TPSAB1 replication number we generate new individualized clinical reference values for the upper limit of ‘normal’. Using this personalized laboratory medicine approach, we demonstrate the clinical utility of tryptase genotyping, finding that in the absence of HαT, BST levels >11.4 ng/mL frequently identify indolent clonal mast cell disease. Moreover, substantial BST elevations (e.g., >100 ng/mL) which would ordinarily prompt bone marrow biopsy, can result from TPSAB1 replications alone and thus be within ‘normal’ limits for certain individuals with HαT.
- Published
- 2022
26. T Cells Expressing a Fully-Human Anti-BCMA Chimeric Antigen Receptor with a Heavy-Chain-Only Antigen-Recognition Domain Exhibit Rapid and Durable Activity Against Multiple Myeloma
- Author
-
Lekha Mikkilineni, Elisabet E. Manasanch, Danielle Natrakul, Katherine Weissler, Jennifer N. Brudno, Jennifer Mann, Stephanie L Goff, James C Yang, Norris Lam, Irina Maric, Hao-Wei Wang, Constance M. Yuan, David F. Stroncek, Steven Highfill, Micaela Ganadan, Rashmika Patel, Steven A Rosenberg, and James N. Kochenderfer
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
27. Eosinophilia in Vexas Syndrome: Expanding Hematologic Phenotype
- Author
-
Ashvind Anand Prabahran, Fernanda Gutierrez-Rodrigues, Serene Ahmad, Edward W Cowen, Ivana Darden, Emma M. Groarke, Marcela Ferrada, Peter C. Grayson, Amy D Klion, Katherine R. Calvo, Irina Maric, Neal S. Young, and Bhavisha A. Patel
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
28. Clinical and biological implications of target occupancy in CLL treated with the BTK inhibitor acalabrutinib
- Author
-
Sarah E. M. Herman, Hailey Harris, Priti Patel, Raquel Izumi, Maryalice Stetler-Stevenson, Constance M. Yuan, Michael Gulrajani, Todd Covey, Irina Maric, Christopher Pleyer, Jean Cheung, Adrian Wiestner, Stefania Pittaluga, Min Hui Wang, Clare Sun, Pia Nierman, Mohammed Farooqui, Ellen K. Kendall, Erika M Gaglione, Inhye E. Ahn, and Ahmed Hamdy
- Subjects
Male ,Chronic lymphocytic leukemia ,Immunology ,Pain ,Phases of clinical research ,Antineoplastic Agents ,Pharmacology ,Biochemistry ,Drug Administration Schedule ,immune system diseases ,hemic and lymphatic diseases ,Agammaglobulinaemia Tyrosine Kinase ,Humans ,Medicine ,Bruton's tyrosine kinase ,RNA, Messenger ,RNA, Neoplasm ,RNA-Seq ,Dosing ,Progression-free survival ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,biology ,business.industry ,Headache ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Hematologic Diseases ,Leukemia, Lymphocytic, Chronic, B-Cell ,Progression-Free Survival ,Neoplasm Proteins ,Leukemia ,Treatment Outcome ,Enzyme Induction ,Pyrazines ,Pharmacodynamics ,Benzamides ,biology.protein ,Acalabrutinib ,Female ,Transcriptome ,business - Abstract
Inhibition of the B-cell receptor pathway, and specifically of Bruton tyrosine kinase (BTK), is a leading therapeutic strategy in B-cell malignancies, including chronic lymphocytic leukemia (CLL). Target occupancy is a measure of covalent binding to BTK and has been applied as a pharmacodynamic parameter in clinical studies of BTK inhibitors. However, the kinetics of de novo BTK synthesis, which determines occupancy, and the relationship between occupancy, pathway inhibition and clinical outcomes remain undefined. This randomized phase 2 study investigated the safety, efficacy, and pharmacodynamics of a selective BTK inhibitor acalabrutinib at 100 mg twice daily (BID) or 200 mg once daily (QD) in 48 patients with relapsed/refractory or high-risk treatment-naïve CLL. Acalabrutinib was well tolerated and yielded an overall response rate (ORR) of partial response or better of 95.8% (95% confidence interval [CI], 78.9-99.9) and an estimated progression-free survival (PFS) rate at 24 months of 91.5% (95% CI, 70.0-97.8) with BID dosing and an ORR of 79.2% (95% CI, 57.9-92.9) and an estimated PFS rate at 24 months of 87.2% (95% CI, 57.2-96.7) with QD dosing. BTK resynthesis was faster in patients with CLL than in healthy volunteers. BID dosing maintained higher BTK occupancy and achieved more potent pathway inhibition compared with QD dosing. Small increments in occupancy attained by BID dosing relative to QD dosing compounded over time to augment downstream biological effects. The impact of BTK occupancy on long-term clinical outcomes remains to be determined. This trial was registered at www.clinicaltrials.gov as #NCT02337829.
- Published
- 2020
- Full Text
- View/download PDF
29. Acute increases in total serum tryptase unassociated with hemodynamic instability in diffuse cutaneous mastocytosis
- Author
-
Seemal F. Awan, Larry B. Schwartz, Irina Maric, Dean D. Metcalfe, and Melody C. Carter
- Subjects
Pulmonary and Respiratory Medicine ,Mastocytosis, Cutaneous ,Mastocytosis, Systemic ,Immunology ,Hemodynamics ,Immunology and Allergy ,Humans ,Tryptases ,Mast Cells ,Mastocytosis - Published
- 2022
30. Oncogenic RAS commandeers amino acid sensing machinery to aberrantly activate mTORC1 in multiple myeloma
- Author
-
Frances A. Tosto, Ping Chen, Dickran Kazandjian, Craig J. Thomas, George E. Wright, Kelli M. Wilson, Thomas Oellerich, James W. Lord, Carleen Klumpp-Thomas, Irina Maric, Grace Smith, Crystal McKnight, Da-Wei Huang, Jan Wisnieski, Xin Yu, Ryan M. Young, Björn Häupl, Michele Ceribelli, Arnold Bolomsky, James Q. Wang, Erin S Beck, Callie K. Van Winkle, Jameson Travers, James D. Phelan, Stefania Pittaluga, and Yandan Yang
- Subjects
Gene isoform ,chemistry.chemical_classification ,MAPK/ERK pathway ,Mitogen-Activated Protein Kinase Kinases ,Multidisciplinary ,Chemistry ,Mutant ,General Physics and Astronomy ,mTORC1 ,General Chemistry ,Mechanistic Target of Rapamycin Complex 1 ,General Biochemistry, Genetics and Molecular Biology ,Amino acid ,Genes, ras ,Mutation ,Cancer research ,Humans ,Protein Isoforms ,Amino acid transporter ,Amino Acids ,Multiple Myeloma ,Gene ,PI3K/AKT/mTOR pathway ,Transcription Factors - Abstract
Oncogenic mutations within the RAS pathway are common in multiple myeloma (MM), an incurable malignancy of plasma cells. However, the mechanisms of pathogenic RAS signaling in this disease remain enigmatic and difficult to inhibit therapeutically. We employed an unbiased proteogenomic approach to dissect RAS signaling in MM by combining genome-wide CRISPR-Cas9 screening with quantitative mass spectrometry focused on RAS biology. We discovered that mutant isoforms of RAS organized a signaling complex with the amino acid transporter, SLC3A2, and MTOR on endolysosomes, which directly activated mTORC1 by co-opting amino acid sensing pathways. MM tumors with high expression of mTORC1-dependent genes were more aggressive and enriched in RAS mutations, and we detected interactions between RAS and MTOR in MM patient tumors harboring mutant RAS isoforms. Inhibition of RAS-dependent mTORC1 activity synergized with MEK and ERK inhibitors to quench pathogenic RAS signaling in MM cells. This study redefines the RAS pathway in MM and provides a mechanistic and rational basis to target this novel mode of RAS signaling.
- Published
- 2022
31. Evidence of an Abnormal Mast Cell Marrow Compartment in Patients with Idiopathic Anaphylaxis
- Author
-
Arnold Kirshenbaum, Irina Maric, Jonathan Lyons, Dean Metcalfe, and Melody Carter
- Subjects
Immunology ,Immunology and Allergy - Published
- 2023
- Full Text
- View/download PDF
32. Mepolizumab is ineffective for treatment of episodic angioedema with eosinophilia in a pilot study
- Author
-
Paneez Khoury, Michelle Makiya, Rodaba Rahim, David Espinoza, Abbie Bowman, Adam Schiffenbauer, Megan Koch, Charles Anderson, Gregory Constantine, Irina Maric, Xiaoping Sun, Thomas Brown, Nicole Holland, JeanAnne Ware, Lauren Wetzler, Michael Fay, and Amy Klion
- Subjects
Immunology ,Immunology and Allergy - Published
- 2023
- Full Text
- View/download PDF
33. Generation of Functionally Competent Eosinophils and Eosinophil Precursors Using an Induced Pluripotent Stem Cell Model
- Author
-
Gregory Constantine, David Espinoza, Elizabeth Swain, Xiaoping Sun, Irina Maric, Gila Idelman, Patricia Fulkerson, Michelle Makiya, Calman Prussin, and Amy Klion
- Subjects
Immunology ,Immunology and Allergy - Published
- 2023
- Full Text
- View/download PDF
34. Carfilzomib, Lenalidomide, and Dexamethasone Followed by Lenalidomide Maintenance for Prevention of Symptomatic Multiple Myeloma in Patients With High-risk Smoldering Myeloma: A Phase 2 Nonrandomized Controlled Trial
- Author
-
Nishant Tageja, Baris Turkbey, Peter L. Choyke, Joseph Roswarski, William D. Figg, Ola Landgren, Constance M. Yuan, Esther Mena, Nisha Patel, Min-Jung Lee, Raul C. Braylan, Elisabet E. Manasanch, Sham Mailankody, Sunmin Lee, Hao-Wei Wang, Ani Petrosyan, Michael Emanuel, Dickran Kazandjian, Mary Kwok, Alina Dulau-Florea, Manisha Bhutani, Weixin Wang, Mark Roschewski, Katherine R. Calvo, Elizabeth M. Hill, Maryalice Stetler-Stevenson, Candis Morrison, Neha Korde, Liza Lindenberg, Seth M. Steinberg, Alexander Dew, Jane B. Trepel, Yong Zhang, and Irina Maric
- Subjects
Adult ,Male ,Smoldering Multiple Myeloma ,Cancer Research ,medicine.medical_specialty ,Dexamethasone ,law.invention ,chemistry.chemical_compound ,Maintenance therapy ,Randomized controlled trial ,law ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Adverse effect ,Lenalidomide ,Multiple myeloma ,Original Investigation ,Aged ,business.industry ,Middle Aged ,medicine.disease ,Carfilzomib ,Minimal residual disease ,Oncology ,chemistry ,Female ,business ,Multiple Myeloma ,Oligopeptides ,medicine.drug - Abstract
Importance High-risk smoldering myeloma has a 5-year risk of progression to symptomatic multiple myeloma of approximately 75%. Treatment with lenalidomide decreases the risk of progression; however, novel triplet regimens are superior, and earlier disease may be more treatment sensitive. Objective To evaluate the use of carfilzomib, lenalidomide, and dexamethasone (KRd) with lenalidomide maintenance therapy as early intervention in high-risk smoldering myeloma and to determine the rates of minimal residual disease (MRD)–negative complete response (CR). Design, Setting, and Participants In this single-arm, single-center, phase 2 nonrandomized controlled trial, responses were evaluated at every cycle during KRd treatment and every 3 cycles subsequently. Bone marrow biopsies and imaging were performed by cycle 8 and then annually. The study enrolled patients from May 29, 2012, to July 23, 2020, at the National Institutes of Health Clinical Center, a highly specialized tertiary cancer center. Patient key eligibility criteria included a diagnosis of high-risk smoldering myeloma based on the Mayo Clinic, Spanish, and/or Rajkumar, Mateos, and Landgren criteria. Interventions Patients received eight 4-week cycles of intravenous carfilzomib 36 mg/m2(first 2 doses, 20 mg/m2), dexamethasone (20 mg, cycles 1-4; 10 mg, cycles 5-8 twice weekly), and lenalidomide 25 mg (days 1-21) followed by twenty-four 28-day cycles of maintenance lenalidomide 10 mg (days 1-21). Stem cell harvest and storage were optional. Main Outcomes and Measures The primary outcome was the MRD-negative CR rate. Key secondary outcomes included duration of MRD-negative CR and progression to multiple myeloma. Results A total of 54 patients (median age, 59 years [range, 40-79 years]; 30 men [55.6%]; and 2 Asian [3.7%], 15 Black [27.8%], 1 Hispanic [1.9%], and 36 White [66.7%] patients) were enrolled, with a median potential follow-up time of 31.9 months (range, 6.7-102.9 months). The MRD-negative CR rate was 70.4% (95% CI, 56.4%-82.0%), with a median sustained duration of 5.5 years (95% CI, 3.7 years to not estimable). The 8-year probability of being free from progression to multiple myeloma was 91.2% (95% CI, 67.4%-97.9%), and no deaths occurred. Nonhematologic grade 3 adverse events occurred in 21 patients (38.9%) and included thromboembolism, rash, and lung infection, with no grade 4 events. Conclusions and Relevance Results of this phase 2 nonrandomized controlled trial suggest that treatment of high-risk smoldering myeloma with novel triplet regimens, such as KRd and lenalidomide maintenance therapy, may alter the natural history of smoldering myeloma by significantly delaying development of end-organ disease. Randomized clinical trials are needed to confirm this favorable benefit-to-risk profile. Trial Registration ClinicalTrials.gov Identifier:NCT01572480
- Published
- 2021
35. Treatment of High Risk (HR) Smoldering Multiple Myeloma (SMM) with Carfilzomib, Lenalidomide, and Dexamethasone (KRd) Followed By Lenalidomide Maintenance (-R): A Phase 2 Clinical and Correlative Study
- Author
-
Irina Maric, Nisha Patel, Manisha Bhutani, Hao-Wei Wang, Mark Roschewski, Michael Emanuel, Constance M. Yuan, Esther Mena, Monica Epstein, Raul C. Braylan, Katherine R. Calvo, Dickran Kazandjian, Maryalice Stetler-Stevenson, Crystal Lu, Liza Lindenberg, Elizabeth M. Hill, Seth M. Steinberg, Candis Morrison, Alina Dulau-Florea, Elisabet E. Manasanch, Nishant Tageja, Peter L. Choyke, Mary L Kwok, Neha Korde, Ola Landgren, William D. Figg, Alexander Dew, Sham Mailankody, Yong Zhang, and Ashley Carpenter
- Subjects
medicine.medical_specialty ,Immunology ,Phases of clinical research ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Carfilzomib ,law.invention ,Clinical trial ,chemistry.chemical_compound ,Randomized controlled trial ,Maintenance therapy ,chemistry ,law ,Internal medicine ,medicine ,Progression-free survival ,Febrile neutropenia ,Lenalidomide ,medicine.drug - Abstract
Background: HR-SMM is a plasma cell disorder with a 5-year risk of progression to symptomatic multiple myeloma (MM) of ~75% without therapy. Early treatment with novel therapies, may decrease the risk of progression and prolong survival as evidenced by studies (Quiredex and ECOG E3A06) comparing lenalidomide ± dexamethasone to observation. Randomized studies in MM have demonstrated that triplet are superior to doublet regimens and whole exome sequencing in HR-SMM suggests a more treatment-sensitive biology. Together, these support our initial pilot study (Korde et al, JAMA Onc 2015) in using effective therapy with KRd-R as early intervention. Given the favorable initial results of our pilot in terms of minimal residual disease (MRD) negativity rates, we designed a single-arm, phase 2 study with the primary objective of determining the rate of MRD negative complete remissions. Herein, we show that rates of MRD negativity are high and they are sustained with the use of KRd-R. Methods: Patients with HR-SMM (Mayo Clinic or PETHEMA models) received eight 28-day cycles of carfilzomib 20/36 mg/m2 IV days 1, 2, 8, 9, 15, 16; lenalidomide 25 mg PO days 1-21, and dexamethasone 20/10 mg days 1, 2, 8, 9, 15, 16, 22, 23. Transplant eligible patients underwent stem cell collection after 4 cycles of KRd and then resumed treatment without an intent for early high-dose melphalan with stem cell support (HDM-ASCT). After 8 cycles of KRd, patients transitioned to receive maintenance therapy with lenalidomide 10 mg PO days 1-21 for 24 additional cycles. Prophylactic antiviral and anticoagulation was mandated for all. MM laboratory evaluations were performed at the start of every cycle during KRd and every 3 cycles during -R and every 3 months during indefinite follow up. Bone marrow biopsies and PET/CTs were performed by the end of cycle 8 induction and then annually indefinitely. The primary objective was to determine the rate of MRD negative remissions by validated multi-color flow cytometry (≤10-5 sensitivity). Key secondary objectives included progression free survival (PFS) to symptomatic myeloma and biochemical progression per IMWG, duration of MRD negativity, overall response rate (ORR), and duration of response. Results: As of 7/15/2020, 52 patients meeting eligibility criteria were enrolled and their demographics and disease characteristics are shown in Table 1. With a median potential follow up of 27.3 months, the primary objective of MRD negative CR rate was 70.2% and the MRD negative ≥VGPR rate was 80.9%, Table 2. The median duration of MRD negativity was 5.5 years with 2 and 5 -year rates of 78% and 55%, respectively. The median time to progression to MM and time to biochemical progression was not reached with 90-month rates of 90% and 78%, respectively - 2 patients progressed to symptomatic MM and 4 patients biochemically. The ORR was 100% and 78% achieved a best response of stringent CR. No deaths occurred. All grade and Grade 3-4 treatment-related adverse events occurred in 90% and 33% of patients, respectively. Grade 3-4 toxicities occurring in >1 patient included neutropenia (19%), lymphopenia (13%), thromboembolism (12%), anemia (8%), rash (8%), leukopenia (6%), lung infection (6%), ALT increase (4%), diarrhea (4%), hyperglycemia (4%), hypophosphatemia (4%), and thrombocytopenia (4%). Other Grade 3-4 adverse events of interest occurring in ≤1 patient included atrial fibrillation, creatinine increase, dyspnea, febrile neutropenia, heart failure, hypertension, and neoplasm. Treatment discontinuation occurred in 4 patients; 3 due to toxicity and 1 due to patient withdrawal. Conclusions: Treatment of HR-SMM with KRd-R to prevent symptomatic MM resulted in an MRD negative CR rate of 70% with a median duration of 5.5 years. At the 5-year landmark, only 10% of patients developed MM which is favorable compared to historical rates with no treatment of ~75%. Alternative approaches using monotherapy lenalidomide (Lonial et al, JCO 2019) resulted in no CRs and a 5-year progression rate of 22% with a treatment discontinuation rate of 51% compared to 7% in our study. More aggressive approaches include GEM-CESAR (Mateos et al, ASH 2019) incorporating HDM-ASCT with KRd-R. Importantly, the rate of MRD negativity reported in GEM-CESAR was 56% compared to 70% in this study. Overall, the benefit compared to risk with KRd in SMM is very favorable. Future randomized trials will be needed to lock in this conclusion. Disclosures Korde: Amgen: Research Funding; Astra Zeneca: Other: Advisory Board. Mailankody:Physician Education Resource: Honoraria; PleXus Communications: Honoraria; Takeda Oncology: Research Funding; Janssen Oncology: Research Funding; Allogene Therapeutics: Research Funding; Juno Therapeutics, a Bristol-Myers Squibb Company: Research Funding. Manasanch:Quest Diagnostics: Research Funding; Sanofi: Research Funding; JW Pharma: Research Funding; Merck: Research Funding; Takeda: Honoraria; GSK: Honoraria; Sanofi: Honoraria; BMS: Honoraria; Adaptive Biotechnologies: Honoraria; Novartis: Research Funding. Bhutani:BMS: Other: Clinical trial funding to institute, Speakers Bureau; Amgen: Speakers Bureau; MedImmune: Other: Clinical Trial Funding to Institute; Janssen: Other: Clinical Trial Funding to Institute; Prothena: Other: Clinical Trial Funding to Institute; Sanofi Genzyme: Consultancy; Takeda: Other: Clinical trial funding to institute, Speakers Bureau. Landgren:Amgen: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Merck: Other; Karyopharma: Research Funding; Merck: Other; Pfizer: Consultancy, Honoraria; Juno: Consultancy, Honoraria; Cellectis: Consultancy, Honoraria; Glenmark: Consultancy, Honoraria, Research Funding; Seattle Genetics: Research Funding; Juno: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Other: Independent Data Monitoring Committees for clinical trials, Research Funding; Karyopharma: Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Glenmark: Consultancy, Honoraria, Research Funding; Takeda: Other: Independent Data Monitoring Committees for clinical trials, Research Funding; Janssen: Consultancy, Honoraria, Other: Independent Data Monitoring Committees for clinical trials, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Binding Site: Consultancy, Honoraria; Takeda: Other: Independent Data Monitoring Committees for clinical trials, Research Funding; BMS: Consultancy, Honoraria; Cellectis: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Binding Site: Consultancy, Honoraria; Adaptive: Consultancy, Honoraria; Seattle Genetics: Research Funding. OffLabel Disclosure: carfilzomib, lenalidomide, and dexamethasone are not approved for smoldering myeloma.
- Published
- 2020
- Full Text
- View/download PDF
36. Platelet-derived growth factor receptor-alpha-positive myeloid neoplasm presenting as eosinophilic gastrointestinal disease
- Author
-
David E. Kleiner, Tom Brown, Amy D. Klion, Gregory M. Constantine, Irina Maric, JeanAnne Ware, Sheila Kumar, Lauren Thumm, and Natasha Kamal
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Receptor, Platelet-Derived Growth Factor alpha ,Platelet-derived growth factor ,Platelet-Derived Growth Factor Receptor Alpha ,Article ,Myeloid Neoplasm ,Enteritis ,chemistry.chemical_compound ,Neoplasms ,Eosinophilia ,Eosinophilic ,medicine ,Humans ,Immunology and Allergy ,Receptors, Platelet-Derived Growth Factor ,Receptor ,business.industry ,medicine.disease ,chemistry ,Gastrointestinal disease ,Gastritis ,medicine.symptom ,business - Published
- 2020
- Full Text
- View/download PDF
37. Dual antibody immunohistochemistry: an efficient and sensitive tool for the detection of residual disease in chronic lymphocytic leukemia
- Author
-
Mohammed Farooqui, Gerald E. Marti, Laura M. Wake, Maryalice Stetler-Stevenson, Adrian Wiestner, Xin Tian, Inhye E. Ahn, and Irina Maric
- Subjects
medicine.medical_specialty ,Pathology ,Histology ,Chronic lymphocytic leukemia ,H&E stain ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Chemoimmunotherapy ,hemic and lymphatic diseases ,Internal medicine ,Biopsy ,medicine ,Hematology ,medicine.diagnostic_test ,biology ,business.industry ,medicine.disease ,Minimal residual disease ,030220 oncology & carcinogenesis ,biology.protein ,Antibody ,CD5 ,business ,030215 immunology - Abstract
Highly effective treatments for chronic lymphocytic leukemia (CLL) have the potential to reduce significant tumor burden to single cells and therefore require sensitive tools to assess for minimal residual disease (MRD) in bone marrow (BM) biopsies. Flow cytometry (FC) is the current gold standard for detection of MRD, but requires a specialized facility, specific antibody panels, collecting hundreds of thousands-to-millions of cells, and personnel with expertise in the analysis and interpretation of FC MRD data, which may not be feasible in many small laboratories. Dual-antibody immunohistochemistry (DA-IHC) can identify abnormal populations better than morphology alone, but its correlation with FC assessment is not known. Our aims are to characterize the efficacy of DA-IHC in assessing BM samples post-treatment and to compare results with FC to evaluate for residual disease in CLL. We collected 2-year post-therapy data from 33 CLL patients on two treatment protocols, chemoimmunotherapy (CIT) and single agent ibrutinib (IB), as well as BMs from 10 healthy volunteers as morphologic controls. BM biopsy specimens were examined for the presence or absence of CLL based on morphologic evidence of lymphoid infiltration and aberrant co-expression of CD5 and PAX5 DA-IHC. FC using a standard CLL antibody panel was performed in parallel. All IB patients had residual disease detected by DA-IHC and FC, although five patients (22%) were morphologically negative by routine hematoxylin and eosin (H&E) stain. Those without overt morphologic evidence of disease showed DA-IHC-positive interstitial single cells and were positive for residual disease by FC. The assessments by DA-IHC and FC were significantly correlated (p = 0.004). Four patients (40%) treated with CIT were morphologically negative by H&E, and two of these had no detectable CLL by either DA-IHC or FC. The other two patients had low-level disease detected by both DA-IHC and FC. The MRD levels identified by DA-IHC were correlated with those by FC (p
- Published
- 2019
- Full Text
- View/download PDF
38. Molecular underpinnings of clinical disparity patterns in African American vs. Caucasian American multiple myeloma patients
- Author
-
Elli Papaemmanuil, Sham Mailankody, Neha Korde, Malin Hultcrantz, Theresia Akhlaghi, Elizabeth M. Hill, Dickran Kazandjian, Evan H. Rustad, Mary Kwok, Ola Landgren, Alex Dew, Irina Maric, and Venkata Yellapantula
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Oncology ,medicine.medical_specialty ,DNA Copy Number Variations ,Somatic cell ,Plasma cell dyscrasia ,medicine.disease_cause ,lcsh:RC254-282 ,Translocation, Genetic ,White People ,Article ,03 medical and health sciences ,Genetic Heterogeneity ,0302 clinical medicine ,Gene Frequency ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Indel ,Multiple myeloma ,Mutation ,business.industry ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Hematology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Black or African American ,030220 oncology & carcinogenesis ,Female ,KRAS ,Hyperdiploidy ,business ,Multiple Myeloma ,030215 immunology - Abstract
Caucasian Americans (CA) compared with African Americans (AA) have a twofold increased incidence of multiple myeloma (MM) and have an earlier age of diagnosis. However, there is sparse information regarding underlying biological differences across racial/ethnic groups. We characterized genetic alterations using a targeted next-generation sequencing assay called myTYPE, developed at MSKCC, allowing capture of somatic mutations, IgH translocations, gains/losses, and hyperdiploidy. Samples were obtained from the NIH Plasma Cell Dyscrasia Racial Disparity Cohort. In total, 68 patient samples were successfully sequenced and manually curated based on well-established databases. Of the 68 patient samples (47 CA, 21 AA), 84% had at least one type of genomic alteration. Importantly, the IgH translocation, t(11;14), was observed more frequently in the AA group (0 vs. 29%, p = 0.001). Known oncogenic somatic non-synonymous mutations were found in 18 genes and indels in 2 genes. KRAS mutations were the most common mutation found in 16% of patients followed by NRAS and BRAF mutations. TP53 somatic mutations appeared to be more common in CA but lacked significance. This proof-of-principle study indicates the presence of varying underlying tumor biology between racial groups and supports the need of future prospective trials to capture these molecular characteristics.
- Published
- 2019
- Full Text
- View/download PDF
39. Detection of paroxysmal nocturnal hemoglobinuria (PNH) in bone marrow aspirates☆
- Author
-
Irina Maric, Katherine R. Calvo, Alina Dulau-Florea, and Raul C. Braylan
- Subjects
biology ,medicine.diagnostic_test ,Chemistry ,Cell ,Hemoglobinuria, Paroxysmal ,Aerolysin ,Hematology ,CD59 ,Flow Cytometry ,medicine.disease ,Molecular biology ,Flow cytometry ,Haematopoiesis ,medicine.anatomical_structure ,Bone Marrow ,hemic and lymphatic diseases ,Paroxysmal nocturnal hemoglobinuria ,medicine ,biology.protein ,Humans ,Bone marrow ,Antibody - Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired condition in which, due to a mutation of the phosphatidylinositol glycan class A gene, hematopoietic cells lack proteins that are normally anchored to the cell surface by glycosylphosphatidylinositol (GPI). Thus, PNH cells show poor expression of surface proteins, such as CD55 and CD59, and dim or absent binding of fluorescently labeled modified aerolysin (FLAER). In clinical diagnostic laboratories, the detection and quantitation of PNH is currently performed by flow cytometry (FC) analysis of peripheral blood (PB) samples. Although PB remains the preferred source of cells for PNH detection, we and other authors have shown that a careful FC analysis of bone marrow (BM) aspirates can provide results for PNH detection and quantitation equivalent to those obtained with PB. Here, we review studies delineating the expression of GPI-anchored proteins and FLAER binding in normal BM cells, and summarize published findings demonstrating the feasibility of identifying and quantitating PNH cells in BM using FLAER as well as antibodies against GPI-anchored proteins. Detection of PNH cells in BM should be useful in patients with unsuspected PNH or with severe cytopenia in whom PB FC analysis may be challenging.
- Published
- 2019
- Full Text
- View/download PDF
40. Remission of indolent systemic mastocytosis in the absence of targeted therapy
- Author
-
Gregory M, Constantine, Dean D, Metcalfe, Cem, Akin, Xiaoping, Sun, Yun, Bai, A Robin, Eisch, Irina, Maric, and Hirsh D, Komarow
- Subjects
Mastocytosis, Systemic ,Humans ,Immunology and Allergy ,Mast Cells ,Mastocytosis - Published
- 2022
- Full Text
- View/download PDF
41. Long term follow-up of a phase II study of cladribine with concurrent rituximab with hairy cell leukemia variant
- Author
-
Hong Zhou, Julie Feurtado, Hao-Wei Wang, Seth M. Steinberg, Chin-Hsien Tai, Mark Raffeld, Constance M. Yuan, Lacey James-Echenique, Evgeny Arons, Raul C. Braylan, Dai Chihara, Liqiang Xi, Alina Dulau-Florea, Katherine R. Calvo, Robert J. Kreitman, Keyur P. Patel, Maryalice Stetler-Stevenson, and Irina Maric
- Subjects
medicine.medical_specialty ,Leukemia, Hairy Cell ,business.industry ,Remission Induction ,Phases of clinical research ,Purine analogue ,Hematology ,Minimal residual disease ,Gastroenterology ,Confidence interval ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Internal medicine ,Medicine ,Cladribine ,Humans ,Rituximab ,Bone marrow ,business ,Hairy Cell Leukemia Variant ,medicine.drug ,Follow-Up Studies - Abstract
Hairy cell leukemia variant (HCLv) responds poorly to purine analogue monotherapy. Rituximab concurrent with cladribine (CDAR) improves response rates, but long-term outcomes are unknown. We report final results of a phase 2 study of CDAR for patients with HCLv. Twenty patients with 0 to 1 prior courses of cladribine and/or rituximab, including 8 who were previously untreated, received cladribine 0.15 mg/kg on days 1 to 5 with 8 weekly rituximab doses of 375 mg/m2 beginning day 1. Patients received a second rituximab course ≥6 months after cladribine, if and when minimal residual disease (MRD) was detected in blood. The complete remission (CR) rate from CDAR was 95% (95% confidence interval, 75-100). Sixteen (80%) of 20 patients (95% confidence interval, 56-94) became MRD negative according to bone marrow at 6 months. The median duration of MRD-negative CR was 70.1 months, and 7 of 16 are still MRD negative up to 120 months. With a median follow-up of 69.7 months, 11 patients received delayed rituximab, and the 5-year progression-free survival (PFS) and overall survival (OS) were 63.3% and 73.9%, respectively. Five patients with TP53 mutations had shorter PFS (median, 36.4 months vs unreached; P = .0024) and OS (median, 52.4 months vs unreached; P = .032). MRD-negative CR at 6 months was significantly associated with longer PFS (unreached vs 17.4 months; P < .0001) and OS (unreached vs 38.2 months; P < .0001). Lack of MRD in blood at 6 months was also predictive of longer PFS and OS (P < .0001). After progression following CDAR, median OS was 29.7 months. CDAR is effective in HCLv, with better outcomes in patients who achieve MRD-negative CR. This trial is registered at www.clinicaltrials.gov as #NCT00923013.
- Published
- 2021
42. Risk-adapted, ofatumumab-based chemoimmunotherapy and consolidation in treatment-naïve chronic lymphocytic leukemia: a phase 2 study
- Author
-
Thomas E. Hughes, Gerald E. Marti, Clare Sun, Ronald P. Taylor, Susan Soto, Adrian Wiestner, Inhye E. Ahn, Irina Maric, Jennifer Lotter, Erika M Gaglione, Constance M. Yuan, Clifton C. Mo, Maryalice Stetler-Stevenson, Laura M Wake, Cydney M. Nichols, Janet Valdez, Christopher Pleyer, Xin Tian, Jeanine Superata, Margaret A. Lindorfer, Mohammed Farooqui, Sarah E. M. Herman, Dennis C Drinkwater, Pia Nierman, and Sanjal Desai
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Trogocytosis ,Chronic lymphocytic leukemia ,Phases of clinical research ,Ofatumumab ,Antibodies, Monoclonal, Humanized ,Article ,Therapy naive ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Chemoimmunotherapy ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,business.industry ,Cytogenetics ,Hematology ,medicine.disease ,Minimal residual disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Treatment Outcome ,chemistry ,030220 oncology & carcinogenesis ,Immunotherapy ,business ,030215 immunology - Abstract
High-risk cytogenetics and minimal residual disease (MRD) after chemoimmunotherapy (CIT) predict unfavorable outcome in chronic lymphocytic leukemia (CLL). This phase 2 study investigated risk-adapted CIT in treatment-naïve CLL (NCT01145209). Patients with high-risk cytogenetics received induction with fludarabine, cyclophosphamide, and ofatumumab. Those without high-risk cytogenetics received fludarabine and ofatumumab. After induction, MRD positive (MRD+) patients received 4 doses of ofatumumab consolidation. MRD negative (MRD−) patients had no intervention. Of 28 evaluable for response, all responded to induction and 10 (36%) achieved MRD−. Two-year progression-free survival (PFS) was 71.4% (CI(95), 56.5–90.3%). There was no significant difference in median PFS between high-risk and standard-risk group. Ofatumumab consolidation didn’t convert MRD+ to MRD−. In MRD+ group, we saw selective loss of CD20 antigens during therapy. In conclusion, risk-adapted CIT is feasible in treatment-naïve CLL. Ofatumumab consolidation didn’t improve depth of response in MRD+ patients. Loss of targetable CD20 likely reduces efficacy of consolidation therapy.
- Published
- 2021
43. Successful pregnancy in the setting of eosinophil depletion by benralizumab
- Author
-
Lauren Wetzler, Tom Brown, Amy D. Klion, Irina Maric, Scott Manetz, Fei Li Kuang, and Elizabeth Battisto
- Subjects
business.industry ,MEDLINE ,Eosinophil ,Successful pregnancy ,Benralizumab ,Antibodies, Monoclonal, Humanized ,Article ,Eosinophils ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Pregnancy ,Immunology ,Immunology and Allergy ,Medicine ,Humans ,Female ,Anti-Asthmatic Agents ,business - Published
- 2020
44. A phase <scp>II</scp> study of ibrutinib and short‐course fludarabine in previously untreated patients with chronic lymphocytic leukemia
- Author
-
Jeanine Superata, Shakuntala Rampertaap, Susan Soto, Adrian Wiestner, Rui Mu, Irina Maric, Constance M. Yuan, Erika M Gaglione, Inhye E. Ahn, Thomas A. Fleisher, Jennifer Lotter, Christopher Pleyer, Xin Tian, Sergio D. Rosenzweig, Maryalice Stetler-Stevenson, Clare Sun, and Stefania Pittaluga
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Chronic lymphocytic leukemia ,Phases of clinical research ,Hematology ,medicine.disease ,Article ,Fludarabine ,Clinical trial ,Leukemia ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Ibrutinib ,medicine ,Short course ,business ,Survival rate ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
45. A randomized double-blind, placebo-controlled study of omalizumab for idiopathic anaphylaxis
- Author
-
Hyejeong Bolan, Irina Maric, Melody C. Carter, Dean D. Metcalfe, Yun Bai, Erica Brittain, Linda M. Scott, Keith Lumbard, and Daly Cantave
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,Immunology ,Placebo-controlled study ,Omalizumab ,Placebo ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Double-Blind Method ,Internal medicine ,Anti-Allergic Agents ,Clinical endpoint ,Hypersensitivity ,Immunology and Allergy ,Medicine ,Humans ,Anaphylaxis ,Aged ,business.industry ,Immunoglobulin E ,Middle Aged ,medicine.disease ,Placebo Effect ,Diagnosis of exclusion ,030104 developmental biology ,medicine.anatomical_structure ,Epinephrine ,030228 respiratory system ,Female ,Bone marrow ,business ,medicine.drug - Abstract
Background Idiopathic anaphylaxis (IA) is a diagnosis of exclusion, thus taking away the option of therapeutic management focused on eliminating the inciting agent. Epinephrine and antihistamines followed by systemic corticosteroids are the mainstays of therapy for acute events. There is no prophylactic therapy that reliably prevents anaphylaxis. Objective We sought to determine the efficacy of omalizumab in the management of patients with frequent episodes of IA in a double-blind, placebo-controlled trial. Methods We prospectively enrolled 19 patients with frequent IA (≥6 episodes/y) who then underwent a medical evaluation that included a serum tryptase determination, mutational analysis for KIT D816V, and bone marrow evaluation to rule out a clonal mast cell disorder. Computer-generated random numbers were provided by the study pharmacist. The primary end point was anaphylactic events in the 6 months after baseline. Sixteen patients completed the primary trial. Results No statistically significant difference was demonstrated between the placebo and treated groups. There was a trend for efficacy in the treatment group, particularly after 60 days. Overall, the safety profile was favorable without long-term side effects. Conclusions Omalizumab was safely administered to a difficult-to-treat patient population with IA. The efficacy results trended modestly in favor of the treatment group, but no statistically significant differences were detected.
- Published
- 2020
46. Randomized Phase II Study of First-Line Cladribine With Concurrent or Delayed Rituximab in Patients With Hairy Cell Leukemia
- Author
-
Liqiang Xi, Robert J. Kreitman, Constance M. Yuan, Hong Zhou, Lacey R. James, Mark Raffeld, Hao-Wei Wang, Seth M. Steinberg, Katherine R. Calvo, Evgeny Arons, Wyndham H. Wilson, Alina Dulau-Florea, Julie Feurtado, Irina Maric, Raul C. Braylan, Dai Chihara, and Maryalice Stetler-Stevenson
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Phases of clinical research ,Purine analogue ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Hairy cell leukemia ,Cladribine ,Aged ,Leukemia, Hairy Cell ,business.industry ,ORIGINAL REPORTS ,Middle Aged ,medicine.disease ,Clinical trial ,Leukemia ,Rituximab ,Female ,business ,medicine.drug - Abstract
PURPOSE Single-agent purine analog, usually cladribine, has been the standard first-line therapy of hairy cell leukemia (HCL) for 30 years. High complete remission (CR) rates often include minimal residual disease (MRD), leading to relapse and repeated treatments. Rituximab can clear MRD, but long-term results are unknown and optimal timing of rituximab undefined. PATIENTS AND METHODS Patients were randomly assigned to first-line cladribine 0.15 mg/kg intravenously days 1-5 with 8 weekly doses of rituximab 375 mg/m2 begun either day 1 (concurrent, CDAR) or ≥ 6 months later (delayed) after detection of MRD in blood. MRD tests included blood and bone marrow (BM) flow cytometry, and BM immunohistochemistry. RESULTS Sixty-eight patients with purine analog-naïve classic HCL were randomly assigned 1:1 to concurrent versus delayed arms. At 6 months after CDAR versus cladribine monotherapy, CR rates were 100% versus 88% ( P = .11), MRD-free CR rates 97% versus 24% ( P < .0001, primary end point), and blood MRD-free rates 100% versus 50% ( P < .0001), respectively. At 96 months median follow-up, 94% versus 12% remained MRD free. Compared with CDAR, delayed rituximab after cladribine achieved lower rate (67% of 21 evaluable patients; P = .0034) and durability ( P = .0081, hazard radio favoring CDAR, 0.094) of MRD-free CR. Nevertheless, 12 patients in the delayed arm remained MRD free when restaged 6-104 (median, 78) months after last delayed rituximab treatment. Compared with cladribine monotherapy, CDAR led to brief grade 3/4 thrombocytopenia (59% v 9%; P < .0001) and platelet transfusions without bleeding (35% v 0%; P = .0002), but higher neutrophil ( P = .017) and platelet ( P = .0015) counts at 4 weeks. CONCLUSION Achieving MRD-free CR of HCL after first-line cladribine is greatly enhanced by concurrent rituximab and less so by delayed rituximab. Longer follow-up will determine if MRD-free survival leads to less need for additional therapy or cure of HCL.
- Published
- 2020
47. Baseline TP53 mutations in Adults with SCD developing Myeloid Malignancy following Hematopoietic Cell Transplantation
- Author
-
Yuesheng Li, Laura W. Dillon, Jack Ghannam, Christopher S. Hourigan, Xin Xu, Irina Maric, Courtney D. Fitzhugh, and Matthew M. Hsieh
- Subjects
Oncology ,Myeloid Malignancy ,medicine.medical_specialty ,Mutation ,Hematopoietic cell ,business.industry ,medicine.medical_treatment ,Immunology ,Myeloproliferative disease ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Tp53 mutation ,medicine.disease_cause ,Biochemistry ,Transplantation ,Internal medicine ,medicine ,business - Published
- 2020
- Full Text
- View/download PDF
48. Quantification of B-cell maturation antigen, a target for novel chimeric antigen receptor T-cell therapy in Myeloma
- Author
-
Irina Maric, David Venzon, David J. Liewehr, Maryalice Stetler-Stevenson, Constance M. Yuan, Dalia A. Salem, and James N. Kochenderfer
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Immunotherapy, Adoptive ,Sensitivity and Specificity ,Article ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Plasma Cell Myeloma ,Biomarkers, Tumor ,medicine ,Humans ,B-Cell Maturation Antigen ,Aged ,medicine.diagnostic_test ,business.industry ,Hematology ,Middle Aged ,Flow Cytometry ,Immunohistochemistry ,Chimeric antigen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Chimeric Antigen Receptor T-Cell Therapy ,Bone marrow ,Multiple Myeloma ,business - Abstract
B-cell maturation antigen (BCMA) is expressed by normal and malignant plasma cells and is targeted via anti-BCMA chimeric antigen receptor T-cell therapy (BCMA CAR T-cell therapy) in plasma cell myeloma (PCM) patients. Surface BCMA expression is required for CAR T-cell binding and killing. We determined the incidence and intensity of expression of BCMA in bone marrow PCM cells using flow cytometry (FC) and immunohistochemistry (IHC). PCM BCMA expression was assessed by FC in 70 patients and in 43 concurrent specimens by IHC. BCMA expression was detected in 94% of patients. FC could assess BCMA expression in all specimens and expression was quantifiable (QuantiBRITE system, BD Biosciences, San Jose, CA) in 89% of cases. Expression was highly variable and could be numerically classified into dim, moderate or bright levels of expression. In the 43 specimens assessed successfully by both IHC and FC, FC showed higher positivity rate (97%) than IHC (72%), indicating that FC is more useful than IHC in detection of BCMA (p = 0.002; McNemar’s test). We conclude that FC is more sensitive than IHC and can be used to objectively quantify BCMA expression by myeloma cells. IHC is primarily useful when there is significant infiltration of the bone marrow by myeloma and is less sensitive with low numbers of myeloma cells. Furthermore, the ability of FC to differentiate between normal and abnormal plasma cells and to quantify BCMA on these cells, makes it a useful and sensitive tool in screening patients for CAR T-cell therapy and for follow-up post therapy.
- Published
- 2018
- Full Text
- View/download PDF
49. Bone Marrow as a Source of Cells for Paroxysmal Nocturnal Hemoglobinuria Detection
- Author
-
Alina Dulau-Florea, Irina Maric, Danielle M. Townsley, Katherine R. Calvo, Chunjie Jiang, Cynthia E. Dunbar, Neal S. Young, Raul C. Braylan, Thomas Winkler, Mariela Monreal, and Elaine K. Jordan
- Subjects
0301 basic medicine ,CD14 ,CD59 ,Paroxysmal nocturnal hemoglobinuria (PNH) ,CD16 ,Flow cytometry ,Fluorescently labeled aerolysin (FLAER) ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Bone marrow ,biology ,medicine.diagnostic_test ,Chemistry ,Original Articles ,General Medicine ,medicine.disease ,Molecular biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Paroxysmal nocturnal hemoglobinuria ,Antibody ,Clone (B-cell biology) - Abstract
Objectives To determine fluorescently labeled aerolysin (FLAER) binding and glycophosphatidylinositol–anchored protein expression in bone marrow (BM) cells of healthy volunteers and patients with paroxysmal nocturnal hemoglobinuria (PNH) detected in peripheral blood (PB); compare PNH clone size in BM and PB; and detect PNH in BM by commonly used antibodies. Methods Flow cytometry analysis of FLAER binding to leukocytes and expression of CD55/CD59 in erythrocytes. Analysis of CD16 in neutrophils and CD14 in monocytes in BM. Results FLAER binds to all normal BM leukocytes, and binding increases with cell maturation. In PNH, lymphocytic clones are consistently smaller than clones of other BM cells. PNH clones are detectable in mature BM leukocytes with high specificity and sensitivity using common antibodies. Conclusions PNH clone sizes measured in mature BM leukocytes and in PB are comparable, making BM suitable for PNH assessment. We further demonstrate that commonly used reagents (not FLAER or CD55/CD59) can reliably identify abnormalities of BM neutrophils and monocytes consistent with PNH cells.
- Published
- 2018
- Full Text
- View/download PDF
50. Treatment of Patients with T Cells Expressing a Fully-Human Anti-BCMA CAR with a Heavy-Chain Antigen-Recognition Domain Caused High Rates of Sustained Complete Responses and Relatively Mild Toxicity
- Author
-
Jennifer N. Brudno, Tyler Lowe, Hao-Wei Wang, Jennifer Mann, Elisabet E. Manasanch, David F. Stroncek, Stephanie L. Goff, James N. Kochenderfer, Lekha Mikkilineni, Constance M. Yuan, Maryalice Stetler-Stevenson, Steven A. Rosenberg, Danielle Natrakul, Micaela Ganadan, Norris Lam, Rashmika Patel, Steven L. Highfill, Irina Maric, and James Chih-Hsin Yang
- Subjects
High rate ,Heavy chain ,Chemistry ,Immunology ,Toxicity ,Cell Biology ,Hematology ,Antigen recognition ,Biochemistry ,Domain (software engineering) ,Cell biology - Abstract
Multiple myeloma (MM) is a malignancy of plasma cells that is nearly always incurable. T cells expressing chimeric antigen receptors (CAR) that target B-cell maturation antigen (BCMA) can recognize and eliminate MM. The murine or other non-human sequences in the single-chain variable fragments (scFv) of many anti-BCMA CARs can elicit recipient immune responses against CAR T cells. We constructed a CAR incorporating an anti-BCMA fully-human heavy-chain variable domain designated FHVH33. FHVH33 lacks the light chain, the artificial linker sequence, and the 2 linker-associated junctions of a scFv, so FHVH33 is smaller than a scFv and is likely to be less immunogenic. The FHVH33-containing CAR utilized in this clinical trial also incorporated a CD8a hinge and transmembrane domain, a 4-1BB domain, and a CD3z domain. The CAR was designated FHVH33-CD8BBZ and was encoded by a gamma-retroviral vector. T cells expressing FHVH33-CD8BBZ were designated FHVH33-T. The FHVH33-T production process was initiated with unsorted peripheral blood mononuclear cells and took 7 days. The treatment protocol was 300 mg/m 2 of cyclophosphamide and 30 mg/m 2 of fludarabine on days -5 to -3 followed by infusion of FHVH33-T on day 0. Twenty-five patients received FHVH33-T infusions. Median age of the treated patients was 62 (range 39-73). Patients received a median of 6 prior lines of therapy (range 3-10). Five dose levels were assessed (Table). Dose level 4, 6x10 6 CAR + T cells/kg was identified as the maximum feasible dose after considering efficacy and manufacturing factors. Twenty-three of 25 patients (92%) obtained objective responses (OR) of partial response (PR) or better. Seventeen patients (68%) attained a best response of stringent complete response (sCR) or very good partial response (VGPR). Thirteen patients have ongoing responses. To date, the median duration of response is 50 weeks for the highest two dose levels. At present, the overall median progression free survival (PFS) is 78 weeks; as responses are ongoing in 13 patients (52%), PFS will likely improve. Nine of 25 patients had extramedullary plasmacytomas at baseline; patients with extramedullary plasmacytomas at baseline were less likely to achieve sCR (P=0.011). All 25 treated patients were evaluable for toxicity. Eighteen patients had grade 1 or 2 cytokine-release syndrome (CRS), and 6 patients had grade 3 CRS. One patient had no CRS. No patients had grade 4 CRS. Five patients received tocilizumab and 4 patients received corticosteroids for CRS. Two of twenty-five patients had grade 3 neurological toxicity possibly attributable to FHVH33-T. No patient had grade 4 neurologic toxicity attributable to CAR T cells. One patient died of influenza pneumonia. We assessed blood CAR+ cells by quantitative PCR. The median peak blood CAR+ cell level was 126.5 cells/µl (range 3-1071 cells/µl), and the median time post-infusion of peak blood CAR + cell levels was 10.5 days (range 7-14). Peak CAR T-cell level was not associated with obtaining a sCR. In contrast, blood CAR+ T cell levels at both 1 and 2 months after infusion were statistically higher for patients obtaining sCR. For the 1-month time-point, blood CAR+ cell levels in cells/mL were 20 for sCR patients and 4 for not sCR patients (P=0.04). Pretreatment serum BCMA was not statistically different when patients obtaining or not obtaining sCR were compared (median serum BCMA in pg/mL: sCR patients 86,243; not sCR patients 261,675, P=0.20). We assessed cell-surface BCMA expression level on MM cells by antibody binding capacity (ABC) flow cytometry. Cell-surface BCMA expression level was not statistically different in sCR versus not sCR patients (median ABC in sites/cell: sCR patients 844; not sCR patients 535, P=0.29). Patients with MM expressing low levels of BCMA obtained durable responses of greater than 2 years duration, which suggests that FHVH33-T can recognize low levels of cell-surface BCMA. Eight patients had extramedullary plasmacytomas at relapse; 4 patients had plasmacytomas biopsied. Two of the biopsied plasmacytomas were BCMA+, and two were BCMA-negative by immunohistochemistry. FHVH33-CD8BBZ CAR T cells caused relatively mild toxicity and a high rate of sCRs in patients with relapsed MM including MM with low cell-surface BCMA expression. Figure 1 Figure 1. Disclosures Brudno: Kyverna Therapeutics: Membership on an entity's Board of Directors or advisory committees. Lam: Kite, a Gilead Company: Patents & Royalties. Kochenderfer: Kite, a Gilead Company: Patents & Royalties: on anti-CD19 CARs, Research Funding; Bristol Myers Squibb: Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.