Bio


Sydney Lu is a hematologist and medical oncologist in the Division of Hematology, Department of Medicine, studying novel therapeutics for challenging cancers and immune disorders.
Sydney's research career started with graduate studies in the laboratory of Dr. Marcel van den Brink at Memorial Sloan Kettering Cancer Center (MSKCC) studying the biology of pathologic donor T cells during graft-versus-host-disease and beneficial T cells mediating graft-versus-tumor effects after allogeneic bone marrow transplant, as well as the role of the thymus in regenerating healthy and protective donor-derived T cells post-transplant.
The direct relevance of these cellular therapies and their immediate translational applicability to patients inspired him to attend medical school at Stanford and further training in hematology and medical oncology at Memorial Sloan Kettering. There, as a fellow and junior faculty member, he studied disordered RNA splicing in cancer in the laboratory of Dr. Omar Abdel-Wahab, with the goal of developing novel drugs targeting RNA splicing. This work has led to observations that targeted degradation of the RNA binding protein RBM39 may be a feasible therapeutic for the treatment of myeloid cancers bearing RNA splicing factor mutations and that pharmacologic RNA splicing inhibition can generate MHC I-presented peptide neoantigens which are exploitable for immunotherapy in model systems.

Sydney's laboratory is broadly interested in studying RNA processing and splicing in the contexts of:
1) normal and pathologic immunity and immunotherapy
2) cancer biology
3) normal and malignant hematopoiesis

Clinical Focus


  • Hematology

Academic Appointments


Honors & Awards


  • Travel Scholarship, American Society for Transplantation and Cellular Therapy (formerly ASBMT) (2006)
  • Travel Scholarship, American Society for Transplantation and Cellular Therapy (formerly ASBMT) (2009)
  • Abstract Achievement Award, American Society of Hematology (2009)
  • Mortimer J. Lacher Fellowship, The Lymphoma Foundation (2017, 2018)
  • Loan Repayment Program, National Institutes of Health (2017)
  • Abstract Achievement Award, American Society of Hematology (2018)
  • Research Award, Aplastic Anemia & Myelodysplastic Syndromes International Foundation (2018)
  • Young Investigator Award, Conquer Cancer Foundation (ASCO) (2018)
  • Lymphoma Research Fellowship, American Association for Cancer Research-Astrazenaca (2018)
  • Portlock Challenge - Robert Hirschhorn Endowment Award, Memorial Sloan Kettering Cancer Center (2019)
  • Physician Scientist Fellowship Award (declined), Doris Duke Charitable Foundation (2019)
  • Research Training Award for Fellows (declined), American Society of Hematology (2019)
  • Career Development Award (Fellow), Leukemia and Lymphoma Society (2019)
  • K08 Mentored Clinical Scientist Researcher Career Development Award, NIH/NCI (2020)
  • Bridge Scholar, Parker Institute for Cancer Immunotherapy (2020)
  • Riney Myeloma Initiative, Paula and Rodger Riney Foundation (2021)
  • Clinical Scientist Development Award, Doris Duke Charitable Foundation (2022)
  • Scholar Award - Jr. Faculty (basic science/translational), American Society of Hematology (2023)

Boards, Advisory Committees, Professional Organizations


  • Member, Sigma Xi (2003 - Present)
  • Member, American Association of Immunologists (2009 - Present)
  • Member, American Society of Hematology (2009 - Present)
  • Member, American Society for Transplantation and Cellular Therapy (formerly ASBMT) (2009 - Present)
  • Member, American Society for Clinical Oncology (2014 - Present)
  • Member, American Association for Cancer Research (2017 - Present)
  • Member, Society for Immunotherapy of Cancer (2021 - Present)

Professional Education


  • Board Certification: American Board of Internal Medicine, Hematology (2020)
  • Board Certification: American Board of Internal Medicine, Medical Oncology (2019)
  • Fellowship, Memorial Sloan Kettering Cancer Center, Hematology and Medical Oncology (2019)
  • Board Certification: American Board of Internal Medicine, Internal Medicine (2016)
  • Residency, New York Presbyterian Hospitals, Weill Cornell Campus, Internal Medicine (2015)
  • Medical Education: Stanford University School of Medicine (2013) CA
  • PhD, Weill Cornell Graduate School of Biomedical Sciences, Immunology (2009)
  • BA, Dartmouth College, Chemistry; Biology (2003)

Stanford Advisees


All Publications


  • Systematic evaluation of AML-associated antigens identifies anti-U5 SNRNP200 therapeutic antibodies for the treatment of acute myeloid leukemia. Nature cancer Knorr, K., Rahman, J., Erickson, C., Wang, E., Monetti, M., Li, Z., Ortiz-Pacheco, J., Jones, A., Lu, S. X., Stanley, R. F., Baez, M., Fox, N., Castro, C., Marino, A. E., Jiang, C., Penson, A., Hogg, S. J., Mi, X., Nakajima, H., Kunimoto, H., Nishimura, K., Inoue, D., Greenbaum, B., Knorr, D., Ravetch, J., Abdel-Wahab, O. 2023

    Abstract

    Despite recent advances in the treatment of acute myeloid leukemia (AML), there has been limited success in targeting surface antigens in AML, in part due to shared expression across malignant and normal cells. Here, high-density immunophenotyping of AML coupled with proteogenomics identified unique expression of a variety of antigens, including the RNA helicase U5 snRNP200, on the surface of AML cells but not on normal hematopoietic precursors and skewed Fc receptor distribution in the AML immune microenvironment. Cell membrane localization of U5 snRNP200 was linked to surface expression of the Fcγ receptor IIIA (FcγIIIA, also known as CD32A) and correlated with expression of interferon-regulated immune response genes. Anti-U5 snRNP200 antibodies engaging activating Fcγ receptors were efficacious across immunocompetent AML models and were augmented by combination with azacitidine. These data provide a roadmap of AML-associated antigens with Fc receptor distribution in AML and highlight the potential for targeting the AML cell surface using Fc-optimized therapeutics.

    View details for DOI 10.1038/s43018-023-00656-2

    View details for PubMedID 37872381

    View details for PubMedCentralID 6769702

  • E7820, an anti-cancer sulfonamide, degrades RBM39 in patients with splicing factor mutant myeloid malignancies: a phase II clinical trial. Leukemia Bewersdorf, J. P., Stahl, M., Taylor, J., Mi, X., Chandhok, N. S., Watts, J., Derkach, A., Wysocki, M., Lu, S. X., Bourcier, J., Hogg, S. J., Rahman, J., Chaudhry, S., Totiger, T. M., Abdel-Wahab, O., Stein, E. M. 2023

    View details for DOI 10.1038/s41375-023-02050-4

    View details for PubMedID 37814121

  • Tracking the Evolution of Therapy-Related Myeloid Neoplasms Using Chemotherapy Signatures. Blood Diamond, B. T., Ziccheddu, B., Maclachlan, K. H., Taylor, J., Boyle, E. M., Arango Ossa, J. E., Jahn, T. J., Affer, M., Totiger, T. M., Coffey, D. G., Chandhok, N., Watts, J. M., Cimmino, L., Lu, S. X., Bolli, N., Bolton, K. L., Landau, H. J., Park, J. H., Ganesh, K., McPherson, A., Sekeres, M. A., Lesokhin, A. M., Chung, D. J., Zhang, Y., Ho, C., Roshal, M., Tyner, J. W., Nimer, S. D., Papaemmanuil, E., Usmani, S. Z., Morgan, G. J., Landgren, O., Maura, F. 2023

    Abstract

    Patients treated with cytotoxic therapies, including autologous stem cell transplantation, are at risk for developing therapy-related myeloid neoplasms (tMN). Pre-leukemic clones (i.e., clonal hematopoiesis; CH) are detectable years before the development of these aggressive malignancies, though the genomic events leading to transformation and expansion are not well-defined. Here, leveraging distinctive chemotherapy-associated mutational signatures from whole-genome sequencing data and targeted sequencing of pre-chemotherapy samples, we reconstruct the evolutionary life-history of 39 therapy-related myeloid malignancies. A dichotomy is revealed, in which neoplasms with evidence of chemotherapy-induced mutagenesis from platinum and melphalan are hypermutated and enriched for complex structural variants (i.e., chromothripsis) while neoplasms with non-mutagenic chemotherapy exposures are genomically similar to de novo acute myeloid leukemia. Using chemotherapy-associated mutational signatures as temporal barcodes linked to a discrete clinical exposure in each patient's life, we estimate that several complex events and genomic drivers are acquired after chemotherapy is administered. For patients with prior multiple myeloma who were treated with high-dose melphalan and autologous stem cell transplantation, we demonstrate that tMN can develop from either a reinfused CH clone that escapes melphalan exposure and is selected following reinfusion, or from TP53-mutant CH that survives direct myeloablative conditioning and acquires melphalan-induced DNA-damage. Overall, we reveal a novel mode of tMN progression that is not reliant on direct mutagenesis or even exposure to chemotherapy. Conversely, for tMN that evolve under the influence of chemotherapy-induced mutagenesis, distinct chemotherapies not only select pre-existing CH, but also promote the acquisition of recurrent genomic drivers.

    View details for DOI 10.1182/blood.2022018244

    View details for PubMedID 36626250

  • Tracking the Evolution of Therapy-Related Myeloid Neoplasms Using Chemotherapy Signatures Ziccheddu, B., Diamond, B., Maclachlan, K. H., Taylor, J., Boyle, E. M., Ossa, J., Jacob, J., Affer, M., Totiger, T. M., Coffey, D., Watts, J., Lu, S. X., Bolli, N., Bolton, K. L., Park, J. H., Landau, H., Ganesh, K., McPherson, A., Sekeres, M. A., Lesokhin, A. M., Chung, D. J., Zhang, Y., Ho, C., Roshal, M., Tyner, J. W., Nimer, S. D., Papaemmanuil, E., Usmani, S., Morgan, G. J., Landgren, O., Maura, F. AMER SOC HEMATOLOGY. 2022: 2088-2090
  • A Phase II Clinical Trial of E7820 for Patients with Relapsed/Refractory Myeloid Malignancies with Mutations in Splicing Factor Genes Bewersdorf, J., Stahl, M. F., Taylor, J., Chandhok, N., Watts, J., Derkach, A., Wysocki, M., Kostantakis, V., Lu, S. X., Bourcier, J., Hogg, S., Totiger, T. M., Abdel-Wahab, O., Stein, E. AMER SOC HEMATOLOGY. 2022: 9065-9067
  • Systematic Evaluation of AML-Associated Antigens Identifies Novel Anti-U5 snRNP200 Therapeutic Antibodies for the Treatment of AML Knorr, K., Erickson, C., Wang, E., Rahman, J., Mi, X., Lu, S., Fox, N., Penson, A., Hogg, S., Stanley, R., Inoue, D., Knorr, D., Ravetch, J., Abdel-Wahab, O. AMER SOC HEMATOLOGY. 2022: 2247-2248
  • Attenuation of Graft-Versus-Host-Disease Via Genetic or Pharmacologic Inhibition of the RNA Splicing Factor RBM39 Bourcier, J., Hogg, S. J., Erickson, C., Singer, M. E., Tittley, S., Ito, T., Egorova, A., Kuttiyara, J., Hanash, A. M., Abdel-Wahab, O., Lu, S. X. AMER SOC HEMATOLOGY. 2022: 1163-1164
  • Waldenström Macroglobulinemia Whole Genome Reveals Prolonged Germinal Center Activity and Late Copy Number Aberrations. Blood advances Maclachlan, K. H., Bagratuni, T., Kastritis, E., Ziccheddu, B., Lu, S. X., Yellapantula, V., Famulare, C. A., Argyropoulos, K. V., Derkach, A., Papaemmanuil, E., Dogan, A., Lesokhin, A. M., Usmani, S. Z., Landgren, O., Palomba, M. L., Maura, F., Dimopoulos, M. A. 2022

    Abstract

    The genomic landscape of Waldenström Macroglobulinemia (WM) is characterized by somatic mutations in MYD88, present from the precursor stages. Using the comprehensive resolution of whole genome sequencing (WGS) in 14 CD19-selected primary WM samples; comparing clonal and subclonal mutations revealed that germinal center mutational signatures SBS9 (poly-eta) and SBS84 (AID) have sustained activity, suggesting that the interaction between WM and the germinal center continues over time. Expanding our cohort size with 33 targeted sequencing samples, we interrogated the WM copy number alteration (CNA) landscape and chronology. Interestingly, CNA prevalence progressively increases in symptomatic WM and relapsed disease when comparing to stable precursor stages, with stable precursors lacking genomic complexity. Two MYD88 wild type WGS contained a clonal gain affecting chromosome 12, which is typically an early event in chronic lymphocytic leukemia. Molecular time analysis demonstrated that both chromosomal 12 gain events occurred early in cancer development while other CNA changes tend to occur later in the disease course and are often subclonal. In summary, WGS analysis in WM allows the demonstration of sustained germinal center activity over time and allows the reconstruction of the temporal evolution of specific genomic features. In addition, our data suggests that, while MYD88-mutations are central to WM clone establishment and can be observed in precursor disease, CNA may contribute to later phases, and may be used as a biomarker for progression risk from precursor conditions to symptomatic disease.

    View details for DOI 10.1182/bloodadvances.2022008876

    View details for PubMedID 36332058

  • Sustained Minimal Residual Disease Negativity in Multiple Myeloma is Associated with Stool Butyrate and Healthier Plant-Based Diets. Clinical cancer research : an official journal of the American Association for Cancer Research Shah, U. A., Maclachlan, K. H., Derkach, A., Salcedo, M., Barnett, K., Caple, J., Blaslov, J., Tran, L., Ciardiello, A., Burge, M., Shekarkhand, T., Adintori, P., Cross, J., Pianko, M. J., Hosszu, K., McAvoy, D., Mailankody, S., Korde, N., Hultcrantz, M., Hassoun, H., Tan, C., Lu, S. X., Patel, D., Diamond, B., Shah, G., Scordo, M., Lahoud, O., Chung, D. J., Landau, H., Usmani, S. Z., Giralt, S., Taur, Y., Landgren, C. O., Block, G., Block, T., Peled, J. U., van den Brink, M. R., Lesokhin, A. M. 2022

    Abstract

    Sustained minimal residual disease (MRD) negativity is associated with long-term survival in multiple myeloma (MM). The gut microbiome is affected by diet, and in turn can modulate host immunity, for example through production of short-chain fatty acids including butyrate. We hypothesized that dietary factors affect the microbiome (abundance of butyrate-producing bacteria or stool butyrate concentration) and may be associated with MM outcomes.We examined the relationship of dietary factors (via a food frequency questionnaire), stool metabolites (via gas chromatography-mass spectrometry), and the stool microbiome (via 16S sequencing - α-diversity and relative abundance of butyrate-producing bacteria) with sustained MRD negativity (via flow cytometry at 2 timepoints 1 year apart) in myeloma patients on lenalidomide maintenance. The Healthy Eating Index 2015 score and flavonoid nutrient values were calculated from the food frequency questionnaire. The Wilcoxon rank sum test was used to evaluate associations with two-sided p < 0.05 considered significant.At 3 months, higher stool butyrate concentration (p=0.037), butyrate producers (p=0.025) and α-diversity (p=0.0035) were associated with sustained MRD-negativity. Healthier dietary proteins, (from seafood and plants), correlated with butyrate at 3 months (p=0.009) and sustained MRD-negativity (p=0.05). Consumption of dietary flavonoids, plant nutrients with antioxidant effects, correlated with stool butyrate concentration (anthocyanidins p=0.01, flavones p=0.01, and flavanols p=0.02).This is the first study to demonstrate an association between a plant-based dietary pattern, stool butyrate production and sustained MRD-negativity in MM; providing rationale to evaluate a prospective dietary intervention.

    View details for DOI 10.1158/1078-0432.CCR-22-0723

    View details for PubMedID 36170461

  • Chemotherapy signatures reveal the evolutionary history of post-melphalan myeloid neoplasm Diamond, B., Ziccheddu, B., Maclachlan, K., Taylor, J., Boyle, E., Coffey, D., Lu, S., Bolli, N., Bolton, K., Park, J., Landau, H., McPherson, A., Sekeres, M., Lesokhin, A., Chung, D., Zhang, Y., Ho, C., Roshal, M., Tyner, J., Nimer, S., Papaemmanuil, E., Usmani, S., Morgan, G., Landgren, O., Maura, F. CIG MEDIA GROUP, LP. 2022: S9
  • Evaluating serum free light chain ratio as a biomarker for multiple myeloma Akhlaghi, T., Maclachlan, K., Korde, N., Mailankody, S., Lesokhin, A., Hassoun, H., Lu, S., Patel, D., Shah, U., Tan, C., Derkach, A., Lahoud, O., Landau, H., Shah, G., Scordo, M., Chung, D., Giralt, S., Usmani, S., Landgren, O., Hultcrantz, M. CIG MEDIA GROUP, LP. 2022: S128-S129
  • African American patients with smoldering multiple myeloma may have a lower risk of progression compared to white patients Akhlaghi, T., Maclachlan, K., Korde, N., Mailankody, S., Lesokhin, A., Hassoun, H., Lu, S., Patel, D., Shah, U., Tan, C., Derkach, A., Lahoud, O., Landau, H., Shah, G., Scordo, M., Chung, D., Giralt, S., Usmani, S., Landgren, O., Hultcrantz, M. CIG MEDIA GROUP, LP. 2022: S129
  • Bone remineralization of lytic lesions in newly diagnosed multiple myeloma (NDMM) patients treated with carfilzomib, lenalidomide, dexamethasone plus /- daratumumab induction regimen Meseha, M., Abusamra, M., Farooki, A., Derkach, A., Maclachlan, K., Hultcrantz, M., Hassoun, H., Shah, U., Lu, S., Mailankody, S., Patel, D., Lahoud, O., Shah, G., Scordo, M., Chung, D., Landau, H., Lesokhin, A., Korde, N., Giralt, S., Usmani, S., Tan, C. CIG MEDIA GROUP, LP. 2022: S139-S140
  • Chemotherapy-related mutational signatures reveal the origins of therapy-related myeloid neoplasms. Diamond, B., Ziccheddu, B., Boyle, E. M., Maclachlan, K., Taylor, J., Watts, J. M., Lu, S. X., Coffey, D. G., Bolli, N., Papaemmanuil, E., Bolton, K., Park, J. H., Landau, H., Ganesh, K., Sekeres, M. A., Nimer, S., Chung, D. J., Ho, C. H., Roshal, M., Lesokhin, A., Morgan, G., Landgren, O., Maura, F. AMER ASSOC CANCER RESEARCH. 2022
  • Chemotherapy-related mutational signatures reveal the origins of therapy-related myeloid neoplasms Diamond, B., Ziccheddu, B., Boyle, E. M., Maclachlan, K., Taylor, J., Watts, J. M., Lu, S. X., Coffey, D. G., Bolli, N., Papaemmanuil, E., Bolton, K., Park, J. H., Landau, H., Ganesh, K., Sekeres, M. A., Nimer, S., Chung, D. J., Ho, C. H., Roshal, M., Lesokhin, A., Morgan, G., Landgren, O., Maura, F. AMER ASSOC CANCER RESEARCH. 2022
  • African American patients with smoldering multiple myeloma may have a lower risk of progression compared to White patients. Akhlaghi, T., Maclachlan, K., Korde, N., Mailankody, S., Lesokhin, A. M., Hassoun, H., Lu, S. X., Patel, D., Shah, U. A., Tan, C., Derkach, A., Lahoud, O., Landau, H., Shah, G. L., Scordo, M., Chung, D. J., Giralt, S., Usmani, S., Landgren, C., Hultcrantz, M. LIPPINCOTT WILLIAMS & WILKINS. 2022
  • Evaluating serum-free light chain ratio as a biomarker for multiple myeloma. Akhlaghi, T., Maclachlan, K., Korde, N., Mailankody, S., Lesokhin, A. M., Hassoun, H., Lu, S. X., Patel, D., Shah, U. A., Tan, C., Derkach, A., Lahoud, O., Landau, H., Shah, G. L., Scordo, M., Chung, D. J., Giralt, S., Usmani, S., Landgren, C., Hultcrantz, M. LIPPINCOTT WILLIAMS & WILKINS. 2022
  • Clinical efficacy of daratumumab (DARA)-based second line therapy after DARAcontaining and DARA-free induction therapies in multiple myeloma: A single center experience. Ciardiello, A., Korde, N., Hultcrantz, M., Hassoun, H., Mailankody, S., Shah, U. A., Lu, S. X., Patel, D., Maclachlan, K., Lahoud, O., Shah, G. L., Scordo, M., Chung, D. J., Landau, H., Giralt, S., Derkach, A., Usmani, S., Lesokhin, A. M., Tan, C. LIPPINCOTT WILLIAMS & WILKINS. 2022
  • Mechanisms of Resistance to Noncovalent Bruton's Tyrosine Kinase Inhibitors. The New England journal of medicine Wang, E., Mi, X., Thompson, M. C., Montoya, S., Notti, R. Q., Afaghani, J., Durham, B. H., Penson, A., Witkowski, M. T., Lu, S. X., Bourcier, J., Hogg, S. J., Erickson, C., Cui, D., Cho, H., Singer, M., Totiger, T. M., Chaudhry, S., Geyer, M., Alencar, A., Linley, A. J., Palomba, M. L., Coombs, C. C., Park, J. H., Zelenetz, A., Roeker, L., Rosendahl, M., Tsai, D. E., Ebata, K., Brandhuber, B., Hyman, D. M., Aifantis, I., Mato, A., Taylor, J., Abdel-Wahab, O. 2022; 386 (8): 735-743

    Abstract

    BACKGROUND: Covalent (irreversible) Bruton's tyrosine kinase (BTK) inhibitors have transformed the treatment of multiple B-cell cancers, especially chronic lymphocytic leukemia (CLL). However, resistance can arise through multiple mechanisms, including acquired mutations in BTK at residue C481, the binding site of covalent BTK inhibitors. Noncovalent (reversible) BTK inhibitors overcome this mechanism and other sources of resistance, but the mechanisms of resistance to these therapies are currently not well understood.METHODS: We performed genomic analyses of pretreatment specimens as well as specimens obtained at the time of disease progression from patients with CLL who had been treated with the noncovalent BTK inhibitor pirtobrutinib. Structural modeling, BTK-binding assays, and cell-based assays were conducted to study mutations that confer resistance to noncovalent BTK inhibitors.RESULTS: Among 55 treated patients, we identified 9 patients with relapsed or refractory CLL and acquired mechanisms of genetic resistance to pirtobrutinib. We found mutations (V416L, A428D, M437R, T474I, and L528W) that were clustered in the kinase domain of BTK and that conferred resistance to both noncovalent BTK inhibitors and certain covalent BTK inhibitors. Mutations in BTK or phospholipase C gamma 2 (PLCgamma2), a signaling molecule and downstream substrate of BTK, were found in all 9 patients. Transcriptional activation reflecting B-cell-receptor signaling persisted despite continued therapy with noncovalent BTK inhibitors.CONCLUSIONS: Resistance to noncovalent BTK inhibitors arose through on-target BTK mutations and downstream PLCgamma2 mutations that allowed escape from BTK inhibition. A proportion of these mutations also conferred resistance across clinically approved covalent BTK inhibitors. These data suggested new mechanisms of genomic escape from established covalent and novel noncovalent BTK inhibitors. (Funded by the American Society of Hematology and others.).

    View details for DOI 10.1056/NEJMoa2114110

    View details for PubMedID 35196427

  • A Pilot Plant-Based Dietary Intervention in Overweight and Obese Patients with Monoclonal Gammopathy of Undetermined Significance and Smoldering Multiple Myeloma- the Nutrition Prevention (NUTRIVENTION) Study Shah, U. A., Alicea, D., Adintori, P. A., Burge, M., Blaslov, J., Derkach, A., Mailankody, S., Korde, N., Tan, C., Hultcrantz, M., Lu, S. X., Patel, D., Hassoun, H., D'Souza, A., Giralt, S. A., Iyengar, N., Landgren, O., van den Brink, M., Lesokhin, A. AMER SOC HEMATOLOGY. 2021
  • Belantamab Mafodotin in Patients with Relapsed/Refractory Multiple Myeloma, a Real-World Experience Hultcrantz, M., Orozco, J., Peterson, T. J., Derkach, A., Hassoun, H., Korde, N., Lu, S. X., Mailankody, S., Patel, D., Shah, U. A., Tan, C., Maclachlan, K. H., Chung, D. J., Landau, H. J., Shah, G. L., Lahoud, O. B., Scordo, M., Landgren, O., Giralt, S., Lesokhin, A. AMER SOC HEMATOLOGY. 2021
  • The Genomic Landscape of Waldenstrom Macroglobulinemia Reveals Sustained Germinal Center Activity and LateDeveloping Copy Number Aberrations Maclachlan, K. H., Bagratuni, T., Kastritis, E., Ziccheddu, B., Lu, S. X., Yellapantula, V. D., Famulare, C., Diamond, B., Chojnacka, M., Raj, A., Mason, E., Coffey, D. G., Hoffman, J. E., Kazandjian, D., Derkach, A., Papaemmanuil, E., Dogan, A., Lesokhin, A., Landgren, O., Palomba, M., Maura, F., Dimopoulos, M. AMER SOC HEMATOLOGY. 2021
  • Daratumumab Versus Lenalidomide Maintenance Therapy for Multiple Myeloma: A Randomized Pilot Study Comparing Patient-Reported Health Related Quality of Life Measures Shah, U. A., Malik, M., Werner, K., Burge, M., Devlin, S. M., Mailankody, S., Korde, N., Hultcrantz, M., Hassoun, H., Tan, C., Maclachlan, K. H., Lu, S. X., Patel, D., Hamilton, A., Scordo, M., Dahi, P. B., Lahoud, O. B., Chung, D. J., Landau, H. J., Shah, G. L., Giralt, S. A., Lesokhin, A., Landgren, O. AMER SOC HEMATOLOGY. 2021
  • Sustained minimal residual disease negativity in Multiple Myeloma is impacted positively by stool butyrate and healthier plant forward diets Shah, U., Derkach, A., Adintori, P., Cross, J., Maclachlan, K., Mailankody, S., Korde, N., Hultcrantz, M., Hassoun, H., Tan, C., Lu, S., Patel, D., Shah, G., Scordo, M., Lahoud, O., Chung, D., Landau, H., Giralt, S., Taur, Y., Landgren, O., Block, T., Peled, J., van den Brink, M., Lesokhin, A. CIG MEDIA GROUP, LP. 2021: S61
  • Comparison of venous thromboembolism incidence in newly diagnosed multiple myeloma patients receiving bortezomib, lenalidomide, dexamethasone (RVD) or carfilzomib, lenalidomide, dexamethasone (KRD) with aspirin or rivaroxaban thromboprophylaxis BRITISH JOURNAL OF HAEMATOLOGY Piedra, K., Peterson, T., Tan, C., Orozco, J., Hultcrantz, M., Hassoun, H., Mailankody, S., Lesokhin, A., Shah, U., Lu, S., Patel, D., Derkach, A., Wilkins, C. R., Korde, N. 2022; 196 (1): 105-109

    Abstract

    Incidence of venous thromboembolism (VTE) varies across different regimens in newly diagnosed multiple myeloma (NDMM) patients. Limited data exist on the use of direct oral anticoagulants as thromboprophylaxis in the setting of haematologic malignancies, specifically multiple myeloma. In this retrospective study of 305 NDMM patients, VTE rates in those treated with carfilzomib, lenalidomide, dexamethasone (KRD) + aspirin (ASA), bortezomib, lenalidomide, dexamethasone (RVD) + ASA, and KRD + rivaroxaban were statistically significant, 16·1%, 4·8%, and 4·8%, respectively. The findings confirm a higher incidence of VTE when using KRD induction compared to RVD induction and reveal that the use of low-dose rivaroxaban thromboprophylaxis can mitigate this risk without an observable increase in bleeding rates.

    View details for DOI 10.1111/bjh.17772

    View details for Web of Science ID 000685674300001

    View details for PubMedID 34396516

  • Pharmacologic modulation of RNA splicing enhances anti-tumor immunity CELL Lu, S. X., De Neef, E., Thomas, J. D., Sabio, E., Rousseau, B., Gigoux, M., Knorr, D. A., Greenbaum, B., Elhanati, Y., Hogg, S. J., Chow, A., Ghosh, A., Xie, A., Zamarin, D., Cui, D., Erickson, C., Singer, M., Cho, H., Wang, E., Lu, B., Durham, B. H., Shah, H., Chowell, D., Gabel, A. M., Shen, Y., Liu, J., Jin, J., Rhodes, M. C., Taylor, R. E., Molina, H., Wolchok, J. D., Merghoub, T., Diaz, L. A., Abdel-Wahab, O., Bradley, R. K. 2021; 184 (15): 4032-+

    Abstract

    Although mutations in DNA are the best-studied source of neoantigens that determine response to immune checkpoint blockade, alterations in RNA splicing within cancer cells could similarly result in neoepitope production. However, the endogenous antigenicity and clinical potential of such splicing-derived epitopes have not been tested. Here, we demonstrate that pharmacologic modulation of splicing via specific drug classes generates bona fide neoantigens and elicits anti-tumor immunity, augmenting checkpoint immunotherapy. Splicing modulation inhibited tumor growth and enhanced checkpoint blockade in a manner dependent on host T cells and peptides presented on tumor MHC class I. Splicing modulation induced stereotyped splicing changes across tumor types, altering the MHC I-bound immunopeptidome to yield splicing-derived neoepitopes that trigger an anti-tumor T cell response in vivo. These data definitively identify splicing modulation as an untapped source of immunogenic peptides and provide a means to enhance response to checkpoint blockade that is readily translatable to the clinic.

    View details for DOI 10.1016/j.cell.2021.05.038

    View details for Web of Science ID 000676120800016

    View details for PubMedID 34171309

    View details for PubMedCentralID PMC8684350

  • Dynamics of minimal residual disease in patients with multiple myeloma on continuous lenalidomide maintenance: a single-arm, single-centre, phase 2 trial LANCET HAEMATOLOGY Diamond, B., Korde, N., Lesokhin, A. M., Smith, E. L., Shah, U., Mailankody, S., Huftcraritz, M., Hassoun, H., Lu, S. X., Tan, C., Rustad, E. H., Maura, F., Maclachlan, K., Peterson, T., Derkach, A., Devlin, S., Landau, H. J., Scordo, M., Chung, D. J., Shah, G. L., Lahoud, O., Thoren, K., Murata, K., Ramanathan, L., Arcila, M. E., Ho, C., Roshal, M., Dogan, A., Giralt, S. A., Landgren, O. 2021; 8 (6): E422-E432

    Abstract

    Background Lenalidomide maintenance improves progression-free survival for patients with multiple myeloma, although its optimal duration is unknown. Clearance of minimal residual disease (MRD) in the bone marrow results in superior outcomes, although its attainment or sustainment does not alter clinical decision-making. Studies that have evaluated MRD serially are limited in length. We therefore aimed to evaluate longitudinal changes in MRD-status (dynamics) and their association with progression-free survival in patients with multiple myeloma.In this single-centre, single-arm, phase 2 study, we enrolled patients aged 18 years and older from the Memorial Sloan Kettering Cancer Center (New York, NY, USA) who had newly diagnosed multiple myeloma following unrestricted frontline therapy and an Eastern Cooperative Oncology Group Performance Status of 2 or lower, including patients who started maintenance before study enrolment. All participants received lenalidomide maintenance at 10 mg for 21 days of 28-day cycles until progression or unacceptable toxic effects for up to 5 years on protocol. The primary endpoint was progression-free survival at 60 months per protocol and key secondary endpoints were MRD rates after completion of the 12th, 24th, and 36th cycle of maintenance and the association between progression-free survival and annual measurement of MRD status. MRD was assessed from first-pull bone marrow aspirates at baseline and annually by flow cytometry per International Myeloma Working Group criteria, (limit of detection of at least 1 × 10-5) up to a maximum of 5 years. Patients who completed at least four cycles of treatment were included in the analysis of the primary endpoint, and patients who had completed at least one dose of treatment on protocol were assessable for secondary endpoints. The study was registered at ClinicalTrials.gov, NCT02538198, and is now closed to accrual.Between Sept 8, 2015, and Jan 25, 2019, 108 patients (100 evaluable for the primary endpoint) were enrolled. Median follow-up was 40·7 months (95% CI 38·7-45·0). At 60 months, progression-free survival was 64% (95% CI 52-79). Median progression-free survival was unreached (95% CI unreached-unreached). MRD dynamics were assessed using 340 MRD assessments done over 5 years for 103 evaluable patients. Patients who sustained MRD negativity for 2 years (n=34) had no recorded disease progression at median 19·8 months (95% CI 15·8-22·3) past the 2-year maintenance landmark. By contrast, patients who lost their MRD-negative responses (n=10) were more likely to progress than those with sustained MRD negativity (HR infinite; p<0·0001) and those with persistent MRD positivity (HR 5·88, 95% CI 1·18-33·33; p=0·015) at the 2-year landmark. Haematological and non-haematological serious adverse events occurred in 19 patients (18%). The most common adverse events of grade 3 or worse were decreased lymphocyte count in 48 (44%) patients and decreased neutrophil count in 47 (44%) patients. One death occurred on study due to sepsis and heart failure and was considered unrelated to the study drug.Serial measurements of MRD allow for dynamic assessment of risk for disease progression. Early intervention should be investigated for patients with loss of MRD negativity. Sustained MRD positivity is not categorically an unfavourable outcome and might portend prolonged stability of low-level disease.Memorial Sloan Kettering and Celgene.

    View details for Web of Science ID 000655263700011

    View details for PubMedID 34048681

  • Using mobile wearables to establish sleep bioprofiles in newly diagnosed multiple myeloma (MM) patients. Hevroni, G., Mastey, D., Tavitian, E., Derkach, A., Salcedo, M., Mailankody, S., Hassoun, H., Lesokhin, A. M., Smith, E. L., Hultcrantz, M., Shah, U. A., Tan, C., Lu, S. X., Shah, G. L., Giralt, S., Devlin, S. M., Atkinson, T., Lengfellner, J. M., Landgren, C., Korde, N. LIPPINCOTT WILLIAMS & WILKINS. 2021
  • Using MALDI-TOF mass spectrometry in peripheral blood for the follow up of newly diagnosed multiple myeloma patients treated with daratumumab-based combination therapy CLINICA CHIMICA ACTA Eveillard, M., Korde, N., Ciardiello, A., Diamond, B., Lesokhin, A., Mailankody, S., Smith, E., Hassoun, H., Hultcrantz, M., Shah, U., Lu, S., Salcedo, M., Werner, K., Rispoli, J., Mastey, D., Landgren, O., Thoren, K. 2021; 516: 136-141

    Abstract

    Daratumumab-based combination therapies have shown high rates of complete response (CR) and minimal residual disease negativity in patients with multiple myeloma. However, daratumumab, an IgGκ monoclonal antibody, interferes with electrophoretic techniques making it difficult to reliably define residual disease versus CR, especially in patients with IgGκ multiple myeloma.Enrichment with polyclonal sheep antibody-coated magnetic microparticles combined with MALDI-TOF mass spectrometry (MALDI-TOF MS) analysis was used to detect M-proteins in serial samples from newly diagnosed multiple myeloma patients treated with daratumumab-based therapy. The performance of the MALDI-TOF MS assay was compared to that of a routine test panel (serum protein electrophoresis (SPEP), immunofixation (IFE) and serum free light chain (FLC)).Comparison of MALDI-TOF MS to SPEP/IFE/FLC showed a concordance of 84.9% (p < 0.001). When MALDI-TOF MS and FLC results were combined, the M-protein detection rate was the same or better than the routine test panel. For the 9 patients who obtained CR during follow-up, MALDI-TOF MS detected an M-protein in 46% of subsequent samples. Daratumumab could be distinguished from the M-protein in 215/222 samples.MALDI-TOF MS is useful in assessing CR in patients treated with monoclonal antibody-based therapies.

    View details for DOI 10.1016/j.cca.2021.01.021

    View details for Web of Science ID 000631257500016

    View details for PubMedID 33545108

    View details for PubMedCentralID PMC7994191

  • Safety and Effectiveness of Weekly Carfilzomib, Lenalidomide, Dexamethasone, and Daratumumab Combination Therapy for Patients With Newly Diagnosed Multiple Myeloma The MANHATTAN Nonrandomized Clinical Trial JAMA ONCOLOGY Landgren, O., Hultcrantz, M., Diamond, B., Lesokhin, A. M., Mailankody, S., Hassoun, H., Tan, C., Shah, U. A., Lu, S. X., Salcedo, M., Werner, K., Rispoli, J., Caple, J., Sams, A., Verducci, D., Jones, K., Concepcion, I., Ciardello, A., Chansakul, A., Schlossman, J., Tavitian, E., Shekarkhand, T., Harrison, A., Piacentini, C., Rustad, E. H., Yellapantula, V., Maclaughlan, K., Maura, F., Landau, H. J., Scordo, M., Chung, D. J., Shah, G., Lahoud, O. B., Thoren, K., Murata, K., Ramanathan, L., Arcila, M. E., Ho, C., Roshal, M., Dogan, A., Derkach, A., Giralt, S. A., Korde, N. 2021; 7 (6): 862-868

    Abstract

    Recently, the benefit of adding daratumumab to the proteasome inhibitor-based, 3-drug combination of bortezomib, lenalidomide, and dexamethasone for patients with newly diagnosed multiple myeloma who underwent high-dose melphalan chemotherapy and autologous hemopoietic cell transplant was assessed. Here, we examine the addition of daratumumab to the second-generation proteasome inhibitor-based, 3-drug combination of carfilzomib, lenalidomide, and dexamethasone.To assess the safety and effectiveness of carfilzomib-lenalidomide-dexamethasone-daratumumab combination therapy for patients with newly diagnosed multiple myeloma, in the absence of high-dose melphalan chemotherapy and autologous hemopoietic cell transplant.Clinical and correlative pilot study at the Memorial Sloan Kettering Cancer Center in New York, New York. Patients with newly diagnosed multiple myeloma were enrolled between October 1, 2018, and November 15, 2019. The median follow-up from start of treatment was 20.3 months (95% CI, 19.2-21.9 months).Eight 28-day cycles with intravenous carfilzomib, 20/56 mg/m2 (days 1, 8, and 15); oral lenalidomide, 25 mg, (days 1-21); dexamethasone, 40 mg weekly, orally or intravenously (cycles 1-4), and 20 mg after cycle 4; and intravenous daratumumab, 16 mg/kg (days 1, 8, 15, and 22 [cycles 1-2]; days 1 and 15 [cycles 3-6]; and day 1 [cycles 7 and 8]).The primary end point was the minimal residual disease (MRD) rate, in the absence of high-dose melphalan chemotherapy and autologous hemopoietic cell transplant. Secondary end points included determining safety and tolerability, evaluating rates of clinical response per the International Myeloma Working Group, and estimating progression-free survival (PFS) and overall survival (OS) rates.Forty-one evaluable patients were enrolled (median age, 59 years; range, 30-70 years); 25 (61%) were female, and 20 (49%) had high-risk multiple myeloma. The primary end point (MRD negativity in the bone marrow; 10-5 sensitivity) was achieved in 29 of 41 patients (71%; 95% CI, 54%-83%), and therefore the trial was deemed successful. Median time to MRD negativity was 6 cycles (range, 1-8 cycles). Secondary end points of the overall response rate and the very good partial response or complete response rate were 100% (41 of 41 patients) and 95% (39 of 41 patients), respectively. At 11 months of the median follow-up, the 1-year PFS rate and the OS rate were 98% (95% CI, 93%-100%) and 100%, respectively. Most common (≥2 patients) grade 3 or 4 adverse events were neutropenia (12 patients [27%]), rash (4 patients [9%]), lung infection (3 patients [7%]), and increased alanine aminotransferase level (2 patients [4%]). There were no deaths.In this nonrandomized clinical trial, carfilzomib-lenalidomide-dexamethasone-daratumumab combination therapy was associated with high rates of MRD negativity in patients with newly diagnosed multiple myeloma and high rates of PFS.

    View details for DOI 10.1001/jamaoncol.2021.0611

    View details for Web of Science ID 000640633800002

    View details for PubMedID 33856405

    View details for PubMedCentralID PMC8050789

  • Minor intron retention drives clonal hematopoietic disorders and diverse cancer predisposition NATURE GENETICS Inoue, D., Polaski, J. T., Taylor, J., Castel, P., Chen, S., Kobayashi, S., Hogg, S. J., Hayashi, Y., Pineda, J., El Marabti, E., Erickson, C., Knorr, K., Fukumoto, M., Yamazaki, H., Tanaka, A., Fukui, C., Lu, S. X., Durham, B. H., Liu, B., Wang, E., Mehta, S., Zakheim, D., Garippa, R., Penson, A., Chew, G., McCormick, F., Bradley, R. K., Abdel-Wahab, O. 2021; 53 (5): 707-+

    Abstract

    Most eukaryotes harbor two distinct pre-mRNA splicing machineries: the major spliceosome, which removes >99% of introns, and the minor spliceosome, which removes rare, evolutionarily conserved introns. Although hypothesized to serve important regulatory functions, physiologic roles of the minor spliceosome are not well understood. For example, the minor spliceosome component ZRSR2 is subject to recurrent, leukemia-associated mutations, yet functional connections among minor introns, hematopoiesis and cancers are unclear. Here, we identify that impaired minor intron excision via ZRSR2 loss enhances hematopoietic stem cell self-renewal. CRISPR screens mimicking nonsense-mediated decay of minor intron-containing mRNA species converged on LZTR1, a regulator of RAS-related GTPases. LZTR1 minor intron retention was also discovered in the RASopathy Noonan syndrome, due to intronic mutations disrupting splicing and diverse solid tumors. These data uncover minor intron recognition as a regulator of hematopoiesis, noncoding mutations within minor introns as potential cancer drivers and links among ZRSR2 mutations, LZTR1 regulation and leukemias.

    View details for DOI 10.1038/s41588-021-00828-9

    View details for Web of Science ID 000639645800002

    View details for PubMedID 33846634

    View details for PubMedCentralID PMC8177065

  • Tailored treatment to MRD response: A phase I/II study for newly diagnosed multiple myeloma patients using high dose twice-weekly carfilzomib (45 and 56 mg/m(2)) in combination with lenalidomide and dexamethasone AMERICAN JOURNAL OF HEMATOLOGY Korde, N., Mastey, D., Tavitian, E., Mailankody, S., Lesokhin, A., Hassoun, H., Smith, E. L., Lendvai, N., Hultcrantz, M., Shah, U., Tan, C., Lu, S., Diamond, B., Salcedo, M., Werner, K., Chung, D. J., Scordo, M., Shah, G. L., Lahoud, O., Landau, H., Arcila, M., Ho, C., Roshal, M., Dogan, A., Derkach, A., Devlin, S. M., Giralt, S. A., Landgren, O. 2021; 96 (6): E193-E196

    View details for DOI 10.1002/ajh.26150

    View details for Web of Science ID 000630545600001

    View details for PubMedID 33661527

    View details for PubMedCentralID PMC8251553

  • Accelerated single cell seeding in relapsed multiple myeloma (vol 11, 3617, 2020) NATURE COMMUNICATIONS Landau, H. J., Yellapantula, V., Diamond, B. T., Rustad, E. H., Maclachlan, K. H., Gundem, G., Medina-Martinez, J., Ossa, J., Levine, M. F., Zhou, Y., Kappagantula, R., Baez, P., Attiyeh, M., Makohon-Moore, A., Zhang, L., Boyle, E. M., Ashby, C., Blaney, P., Patel, M., Zhang, Y., Dogan, A., Chung, D. J., Giralt, S., Lahoud, O. B., Peled, J. U., Scordo, M., Shah, G., Hassoun, H., Korde, N. S., Lesokhin, A. M., Lu, S., Mailankody, S., Shah, U., Smith, E., Hultcrantz, M. L., Ulaner, G. A., van Rhee, F., Morgan, G. J., Landgren, O., Papaemmanuil, E., Iacobuzio-Donahue, C., Maura, F. 2021; 12 (1): 591

    View details for DOI 10.1038/s41467-021-20978-y

    View details for Web of Science ID 000613568700002

    View details for PubMedID 33473129

    View details for PubMedCentralID PMC7817844

  • COVID-19 Infections and Clinical Outcomes in Patients with Multiple Myeloma in New York City: A Cohort Study from Five Academic Centers BLOOD CANCER DISCOVERY Hultcrantz, M., Richter, J., Rosenbaum, C. A., Patel, D., Smith, E. L., Korde, N., Lu, S. X., Mailankody, S., Shah, U. A., Lesokhin, A. M., Hassoun, H., Tan, C., Maura, F., Derkach, A., Diamond, B., Rossi, A., Pearse, R. N., Madduri, D., Chari, A., Kaminetzky, D., Braunstein, M. J., Gordillo, C., Reshef, R., Taur, Y., Davies, F. E., Jagannath, S., Niesvizky, R., Lentzsch, S., Morgan, G. J., Landgren, O. 2020; 1 (3): 234-243

    Abstract

    Patients with multiple myeloma have a compromised immune system, due to both the disease and antimyeloma therapies, and may therefore be particularly susceptible to COVID-19. Here, we report outcomes and risk factors for serious disease in patients with multiple myeloma treated at five large academic centers in New York City in the spring of 2020, during which it was a global epicenter of the SARS-CoV-2 pandemic. Of 100 patients with multiple myeloma (male 58%; median age 68) diagnosed with COVID-19, 75 were admitted; of these, 13 patients (17%) were placed on invasive mechanical ventilation, and 22 patients (29%) expired. Of the 25 nonadmitted patients, 4 were asymptomatic. There was a higher risk of adverse outcome (intensive care unit admission, mechanical ventilation, or death) in Hispanics/Latinos (n = 21), OR = 4.7 (95% confidence interval, 1.3-16.7), and African American Blacks (n = 33), OR = 3.5 (1.1-11.5), as compared with White patients (n = 36). Patients who met the adverse combined endpoint had overall higher levels of inflammatory markers and cytokine activation. None of the other studied risk factors were significantly associated (P > 0.05) with adverse outcome: hypertension (n = 56), OR = 2.2 (0.9-5.4); diabetes (n = 18), OR = 0.9 (0.3-2.9); age >65 years (n = 63), OR = 1.8 (0.7-4.6); high-dose melphalan with autologous stem cell transplant <12 months (n = 7), OR = 0.9 (0.2-5.4); and immunoglobulin G <650 mg/dL (n = 42), OR = 0.9 (0.3-2.2). In this largest cohort to date of patients with multiple myeloma and COVID-19, we found the case fatality rate to be 29% among hospitalized patients and that race/ethnicity was the most significant risk factor for adverse outcome.Patients with multiple myeloma are immunocompromised, raising the question whether they are at higher risk of severe COVID-19 disease. In this large case series on COVID-19 in patients with multiple myeloma, we report 29% mortality rates among hospitalized patients and identify race/ethnicity as the most significant risk factor for severe outcome.See related commentary by Munshi and Anderson, p. 218. This article is highlighted in the In This Issue feature, p. 215.

    View details for DOI 10.1158/2643-3230.BCD-20-0102

    View details for Web of Science ID 000713094100004

    View details for PubMedID 34651141

    View details for PubMedCentralID PMC7668224

  • Accelerated single cell seeding in relapsed multiple myeloma NATURE COMMUNICATIONS Landau, H. J., Yellapantula, V., Diamond, B. T., Rustad, E. H., Maclachlan, K. H., Gundem, G., Medina-Martinez, J., Ossa, J., Levine, M. F., Zhou, Y., Kappagantula, R., Baez, P., Attiye, M., Makohon-Moore, A., Zhang, L., Boyle, E. M., Ashby, C., Blaney, P., Patel, M., Zhang, Y., Dogan, A., Chung, D. J., Giralt, S., Lahoud, O. B., Peled, J. U., Scordo, M., Shah, G., Hassoun, H., Korde, N. S., Lesokhin, A. M., Lu, S., Mailankody, S., Shah, U., Smith, E., Hultcrantz, M. L., Ulaner, G. A., van Rhee, F., Morgan, G. J., Landgren, O., Papaemmanuil, E., Iacobuzio-Donahue, C., Maura, F. 2020; 11 (1): 3617

    Abstract

    Multiple myeloma (MM) progression is characterized by the seeding of cancer cells in different anatomic sites. To characterize this evolutionary process, we interrogated, by whole genome sequencing, 25 samples collected at autopsy from 4 patients with relapsed MM and an additional set of 125 whole exomes collected from 51 patients. Mutational signatures analysis showed how cytotoxic agents introduce hundreds of unique mutations in each surviving cancer cell, detectable by bulk sequencing only in cases of clonal expansion of a single cancer cell bearing the mutational signature. Thus, a unique, single-cell genomic barcode can link chemotherapy exposure to a discrete time window in a patient's life. We leveraged this concept to show that MM systemic seeding is accelerated at relapse and appears to be driven by the survival and subsequent expansion of a single myeloma cell following treatment with high-dose melphalan therapy and autologous stem cell transplant.

    View details for DOI 10.1038/s41467-020-17459-z

    View details for Web of Science ID 000552423000036

    View details for PubMedID 32680998

    View details for PubMedCentralID PMC7368016

  • Comparison of MALDI-TOF mass spectrometry analysis of peripheral blood and bone marrow-based flow cytometry for tracking measurable residual disease in patients with multiple myeloma BRITISH JOURNAL OF HAEMATOLOGY Eveillard, M., Rustad, E., Roshal, M., Zhang, Y., Ciardiello, A., Korde, N., Hultcrantz, M., Lu, S., Shah, U., Hassoun, H., Smith, E., Lesokhin, A., Mailankody, S., Landgren, O., Thoren, K. 2020; 189 (5): 904-907

    Abstract

    Matrix-assisted laser desorption ionisation time-of-flight mass spectrometry (MALDI-TOF MS) may soon replace routine electrophoretic methods for monitoring monoclonal proteins in patients with multiple myeloma. To further evaluate the clinical utility of this assay, we compared the performance of MALDI-TOF-MS head-to-head with an established bone marrow-based measurable residual disease assay by flow cytometry (Flow-BM-MRD), using Memorial Sloan Kettering Cancer Center's 10-color, single-tube method. Our results suggest that MALDI-TOF-MS adds value to bone marrow-based MRD testing and may be most useful for early detection of relapse in peripheral blood compared to current electrophoretic methods.

    View details for DOI 10.1111/bjh.16443

    View details for Web of Science ID 000537904500028

    View details for PubMedID 32026474

    View details for PubMedCentralID PMC7275888

  • Modern treatments and future directions for newly diagnosed multiple myeloma patients BEST PRACTICE & RESEARCH CLINICAL HAEMATOLOGY Lu, S. X. 2020; 33 (1): 101151

    Abstract

    Over the course of the past decade-plus, the therapy of newly diagnosed multiple myeloma has seen incredible advances in the domains of diagnostic evaluation, active medical therapy, and response evaluation. This manuscript reviews the evaluation and management of newly diagnosed active multiple myeloma, with a focus on major clinical trials and IMWG recommendations. The paper describes a current approach for the initial evaluation and workup for patients with putative active myeloma, with consideration towards potential MRD-directed therapeutic approaches and future clinical trials, and then discusses management with a focus on induction regimens with attention primarily to modern three and four-drug combinations for transplant-eligible and transplant-ineligible patients, and those with organ dysfunction. Finally, this article briefly reviews minimal residual disease directed therapy approaches, primarily in the context of whether eligible patients should be referred for high dose chemotherapy and autologous stem cell rescue. Maintenance therapy for both transplant eligible and ineligible patients is discussed elsewhere in this issue.

    View details for DOI 10.1016/j.beha.2020.101151

    View details for Web of Science ID 000528536400011

    View details for PubMedID 32139016

  • NOVEL ALTERATIONS IN CSF1R, RET, AND OTHER DIVERSE KINASES IN THE HISTIOCYTOSES WITH BIOCHEMICAL AND STRUCTURAL INSIGHTS INTO THEIR MECHANISMS OF ACTIVATION Durham, B., Lopez-Rodrigo, E., Picarsic, J., Abramson, D., Rotemberg, V., De Munck, S., Pannecoucke, E., Lu, S., Pastore, A., Mandelker, D., Birsoy, O., Ulaner, G., Walsh, M., Yabe, M., Petrova-Drus, K., Arcila, M., Ladanyi, M., Solit, D., Berger, M., Hyman, D., Lacouture, M., Ki, M., Dunkel, I., Lopez, V., Mora, J., Haroche, J., Emile, J., Decaux, O., Geissmann, F., Savvides, S., Drilon, A., Diamond, E., Abdel-Wahab, O. WILEY. 2020: S8-S9
  • Activating mutations in CSF1R and additional receptor tyrosine kinases in histiocytic neoplasms NATURE MEDICINE Durham, B. H., Rodrigo, E., Picarsic, J., Abramson, D., Rotemberg, V., De Munck, S., Pannecoucke, E., Lu, S. X., Pastore, A., Yoshimi, A., Mandelker, D., Ceyhan-Birsoy, O., Ulaner, G. A., Walsh, M., Yabe, M., Petrova-Drus, K., Arcila, M. E., Ladanyi, M., Solit, D. B., Berger, M. F., Hyman, D. M., Lacouture, M. E., Erickson, C., Saganty, R., Ki, M., Dunkel, I. J., Lopez, V., Mora, J., Haroche, J., Emile, J., Decaux, O., Geissmann, F., Savvides, S. N., Drilon, A., Diamond, E. L., Abdel-Wahab, O. 2019; 25 (12): 1839-1842

    Abstract

    Histiocytoses are clonal hematopoietic disorders frequently driven by mutations mapping to the BRAF and MEK1 and MEK2 kinases. Currently, however, the developmental origins of histiocytoses in patients are not well understood, and clinically meaningful therapeutic targets outside of BRAF and MEK are undefined. In this study, we uncovered activating mutations in CSF1R and rearrangements in RET and ALK that conferred dramatic responses to selective inhibition of RET (selpercatinib) and crizotinib, respectively, in patients with histiocytosis.

    View details for DOI 10.1038/s41591-019-0653-6

    View details for Web of Science ID 000500824900026

    View details for PubMedID 31768065

    View details for PubMedCentralID PMC6898787

  • VTE Rates and Safety Analysis of Newly Diagnosed Multiple Myeloma Patients Receiving Carfilzomib-Lenalidomide-Dexamethasone (KRD) with or without Rivaroxaban Prophylaxis Piedra, K. M., Hassoun, H., Buie, L. W., Devlin, S. M., Flynn, J., Hultcrantz, M., Lesokhin, A. M., Lu, S. X., Mailankody, S., Shah, U. A., Smith, E. L., Landgren, O., Korde, N., Peterson, T. J. AMER SOC HEMATOLOGY. 2019
  • Weekly Carfilzomib, Lenalidomide, Dexamethasone and Daratumumab (wKRd-D) Combination Therapy Provides Unprecedented MRD Negativity Rates in Newly Diagnosed Multiple Myeloma: A Clinical and Correlative Phase 2 Study Landgren, O., Hultcrantz, M., Lesokhin, A. M., Mailankody, S., Hassoun, H., Smith, E. L., Shah, U. A., Lu, S. X., Mastey, D., Salcedo, M., Diab, V., Werner, K., Rispoli, J., Sams, A., Verducci, D., Jones, K., Harrison, A., Chansakul, A., Rustad, E. H., Yellapantula, V., Maura, F., Landau, H. J., Scordo, M., Chung, D. J., Shah, G., Lahoud, O. B., Thoren, K., Murata, K., Ramanathan, L., Arcila, M. E., Ho, C., Roshal, M., Dogan, A., Devlin, S. M., Giralt, S., Korde, N. AMER SOC HEMATOLOGY. 2019
  • Spliceosomal Disruption of the Non-Canonical SWI/SNF Chromatin Remodeling Complex in SF3B1 Mutant Leukemias Inoue, D., Guo-Liang, C., Liu, B., Lee, S. C., Michel, B. C., Pangallo, J., D'Avino, A. R., North, K., Bitner, L. E., Xiao, M., Kobayashi, S., Yoshimi, A., Cho, H., Lu, S. X., Taylor, J., Kadoch, C., Bradley, R. K., Abdel-Wahab, O. AMER SOC HEMATOLOGY. 2019
  • Long-Term Sustained Minimal Residual Disease (MRD) Negativity in Multiple Myeloma Patients Treated with Lenalidomide Maintenance Therapy: A Clinical and Correlative Phase 2 Study Landgren, O., Mastey, D., Lesokhin, A. M., Smith, E. L., Shah, U. A., Mailankody, S., Hultcrantz, M., Hassoun, H., Lu, S. X., Salcedo, M., Diab, V., Werner, K., Rispoli, J., Sams, A., Verducci, D., Jones, K., Schlossman, J., Chansakul, A., Harrison, A., Ciardiello, A., Tavitian, E., Shekarkhand, T., Rustad, E., Yellapantula, V., Maura, F., Peterson, T. J., Devlin, S. M., Landau, H. J., Scordo, M., Chung, D. J., Shah, G., Lahoud, O. B., Thoren, K., Murata, K., Ramanathan, L., Arcila, M. E., Ho, C., Roshal, M., Dogan, A., Giralt, S., Korde, N. AMER SOC HEMATOLOGY. 2019
  • MALDI-TOF Mass Spectrometry in Serum for the Follow-up of Newly Diagnosed Multiple Myeloma Patients Treated with Daratumumab-Based Combination Therapy Eveillard, M., Hultcrantz, M., Lesokhin, A. M., Mailankody, S., Smith, E. L., Hassoun, H., Shah, U. A., Lu, S. X., Korde, N., Landgren, O., Thoren, K. AMER SOC HEMATOLOGY. 2019
  • An Observational, Retrospective Analysis of Retreatment with Carfilzomib in the Management of Patients with Multiple Myeloma Peterson, T. J., Korde, N., Hassoun, H., Mailankody, S., Lesokhin, A. M., Hultcrantz, M., Shah, U. A., Smith, E. L., Lu, S. X., Landau, H. J., Shah, G. L., Scordo, M., Lahoud, O. B., Giralt, S. A., Landgren, O. AMER SOC HEMATOLOGY. 2019
  • Spliceosomal disruption of the non-canonical BAF complex in cancer NATURE Inoue, D., Chew, G., Liu, B., Michel, B. C., Pangallo, J., D'Avino, A. R., Hitchman, T., North, K., Lee, S., Bitner, L., Block, A., Moore, A. R., Yoshimi, A., Escobar-Hoyos, L., Cho, H., Penson, A., Lu, S. X., Taylor, J., Chen, Y., Kadoch, C., Abdel-Wahab, O., Bradley, R. K. 2019; 574 (7778): 432-+

    Abstract

    SF3B1 is the most commonly mutated RNA splicing factor in cancer1-4, but the mechanisms by which SF3B1 mutations promote malignancy are poorly understood. Here we integrated pan-cancer splicing analyses with a positive-enrichment CRISPR screen to prioritize splicing alterations that promote tumorigenesis. We report that diverse SF3B1 mutations converge on repression of BRD9, which is a core component of the recently described non-canonical BAF chromatin-remodelling complex that also contains GLTSCR1 and GLTSCR1L5-7. Mutant SF3B1 recognizes an aberrant, deep intronic branchpoint within BRD9 and thereby induces the inclusion of a poison exon that is derived from an endogenous retroviral element and subsequent degradation of BRD9 mRNA. Depletion of BRD9 causes the loss of non-canonical BAF at CTCF-associated loci and promotes melanomagenesis. BRD9 is a potent tumour suppressor in uveal melanoma, such that correcting mis-splicing of BRD9 in SF3B1-mutant cells using antisense oligonucleotides or CRISPR-directed mutagenesis suppresses tumour growth. Our results implicate the disruption of non-canonical BAF in the diverse cancer types that carry SF3B1 mutations and suggest a mechanism-based therapeutic approach for treating these malignancies.

    View details for DOI 10.1038/s41586-019-1646-9

    View details for Web of Science ID 000490988300071

    View details for PubMedID 31597964

    View details for PubMedCentralID PMC6858563

  • Association of Immune Marker Changes With Progression of Monoclonal Gammopathy of Undetermined Significance to Multiple Myeloma JAMA ONCOLOGY Landgren, O., Hofmann, J. N., McShane, C. M., Santo, L., Hultcrantz, M., Korde, N., Mailankody, S., Kazandjian, D., Murata, K., Thoren, K., Ramanathan, L., Dogan, A., Rustad, E., Lu, S. X., Akhlaghi, T., Kristinsson, S. Y., Bjorkholm, M., Devlin, S., Purdue, M. P., Pfeiffer, R. M., Turesson, I. 2019; 5 (9): 1293-1301

    Abstract

    Multiple myeloma is consistently preceded by monoclonal gammopathy of undetermined significance (MGUS). Risk models that estimate the risk of progression from MGUS to multiple myeloma use data from a single time point, usually the initial workup.To longitudinally investigate the alterations of serum immune markers with stable vs progressive MGUS.This prospective cross-sectional cohort study included 77 469 adult participants aged 55 to 74 years in the screening arm of the National Cancer Institute Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial who had a diagnosis of progressing MGUS (n = 187) or stable MGUS (n = 498), including light-chain subtype, from November 1993, through December 2011. For each participant, all available serially stored prediagnostic serum samples (N = 3266) were obtained. Data analysis was performed from April 2018, to December 2018.Serum protein and monoclonal immunoglobulin levels, serum free light chains, and serum light chains within each immunoglobulin class were measured.Of 685 individuals included in the study, 461 (67.3%) were men; the mean (SD) age was 69.1 (5.6) years. In cross-sectional modeling, risk factors associated with progressive MGUS were IgA isotype (adjusted odds ratio [OR], 1.80; 95% CI, 1.03-3.13; P = .04), 15 g/L or more monoclonal spike (adjusted OR, 23.5; 95% CI, 8.9-61.9; P < .001), skewed (<0.1 or >10) serum free light chains ratio (adjusted OR, 46.4; 95% CI, 18.4-117.0; P < .001), and severe immunoparesis (≥2 suppressed uninvolved immunoglobulins) (adjusted OR, 19.1; 95% Cl, 7.5-48.3; P < .001). Risk factors associated with progressive light-chain MGUS were skewed serum free light chains ratio (adjusted OR, 44.0; 95% CI, 14.2-136.3; P < .001) and severe immunoparesis (adjusted OR, 48.6; 95% CI, 9.5-248.2; P < .001). In longitudinal analysis of participants with serial samples prior to progression, 23 of 43 participants (53%) had high-risk MGUS before progression; 16 of these 23 (70%) experienced conversion from low-risk or intermediate-risk MGUS within 5 years. Similar results were found for light-chain MGUS.The findings of evolving risk patterns support annual blood testing and risk assessment for patients with MGUS or light-chain MGUS.

    View details for DOI 10.1001/jamaoncol.2019.1568

    View details for Web of Science ID 000486897100009

    View details for PubMedID 31318385

    View details for PubMedCentralID PMC6646992

  • Corrupted coordination of epigenetic modifications leads to diverging chromatin states and transcriptional heterogeneity in CLL NATURE COMMUNICATIONS Pastore, A., Gaiti, F., Lu, S. X., Brand, R. M., Kulm, S., Chaligne, R., Gu, H., Huang, K. Y., Stamenova, E. K., Beguelin, W., Jiang, Y., Schulman, R. C., Kim, K., Alonso, A., Allan, J. N., Furman, R. R., Gnirke, A., Wu, C. J., Melnick, A. M., Meissner, A., Bernstein, B. E., Abdel-Wahab, O., Landau, D. A. 2019; 10: 1874

    Abstract

    Cancer evolution is fueled by epigenetic as well as genetic diversity. In chronic lymphocytic leukemia (CLL), intra-tumoral DNA methylation (DNAme) heterogeneity empowers evolution. Here, to comprehensively study the epigenetic dimension of cancer evolution, we integrate DNAme analysis with histone modification mapping and single cell analyses of RNA expression and DNAme in 22 primary CLL and 13 healthy donor B lymphocyte samples. Our data reveal corrupted coherence across different layers of the CLL epigenome. This manifests in decreased mutual information across epigenetic modifications and gene expression attributed to cell-to-cell heterogeneity. Disrupted epigenetic-transcriptional coordination in CLL is also reflected in the dysregulation of the transcriptional output as a function of the combinatorial chromatin states, including incomplete Polycomb-mediated gene silencing. Notably, we observe unexpected co-mapping of typically mutually exclusive activating and repressing histone modifications, suggestive of intra-tumoral epigenetic diversity. Thus, CLL epigenetic diversification leads to decreased coordination across layers of epigenetic information, likely reflecting an admixture of cells with diverging cellular identities.

    View details for DOI 10.1038/s41467-019-09645-5

    View details for Web of Science ID 000465201500007

    View details for PubMedID 31015400

    View details for PubMedCentralID PMC6478836

  • Targeting an RNA-Binding Protein Network in Acute Myeloid Leukemia CANCER CELL Wang, E., Lu, S. X., Pastore, A., Chen, X., Imig, J., Lee, S., Hockemeyer, K., Ghebrechristos, Y. E., Yoshimi, A., Inoue, D., Ki, M., Cho, H., Bitner, L., Kloetgen, A., Lin, K., Uehara, T., Owa, T., Tibes, R., Krainer, A. R., Abdel-Wahab, O., Aifantis, I. 2019; 35 (3): 369-+

    Abstract

    RNA-binding proteins (RBPs) are essential modulators of transcription and translation frequently dysregulated in cancer. We systematically interrogated RBP dependencies in human cancers using a comprehensive CRISPR/Cas9 domain-focused screen targeting RNA-binding domains of 490 classical RBPs. This uncovered a network of physically interacting RBPs upregulated in acute myeloid leukemia (AML) and crucial for maintaining RNA splicing and AML survival. Genetic or pharmacologic targeting of one key member of this network, RBM39, repressed cassette exon inclusion and promoted intron retention within mRNAs encoding HOXA9 targets as well as in other RBPs preferentially required in AML. The effects of RBM39 loss on splicing further resulted in preferential lethality of spliceosomal mutant AML, providing a strategy for treatment of AML bearing RBP splicing mutations.

    View details for DOI 10.1016/j.ccell.2019.01.010

    View details for Web of Science ID 000461697400006

    View details for PubMedID 30799057

    View details for PubMedCentralID PMC6424627

  • Dependency of Spliceosomal Mutant MDS on Innate Immune Signaling Lee, S. C., North, K., Chen, C., Kim, E., Lu, S. X., Ki, M., Garippa, R., Lowe, S. W., Bradley, R. K., Abdel-Wahab, O. I. AMER SOC HEMATOLOGY. 2018
  • Synthetic Lethal and Convergent Biological Effects of Cancer-Associated Spliceosomal Gene Mutations CANCER CELL Lee, S., North, K., Kim, E., Jang, E., Obeng, E., Lu, S. X., Liu, B., Inoue, D., Yoshimi, A., Ki, M., Yeo, M., Zhang, X., Kim, M., Cho, H., Chung, Y., Taylor, J., Durham, B. H., Kim, Y., Pastore, A., Monette, S., Palacino, J., Seiler, M., Buonamici, S., Smith, P. G., Ebert, B. L., Bradley, R. K., Abdel-Wahab, O. 2018; 34 (2): 225-+

    Abstract

    Mutations affecting RNA splicing factors are the most common genetic alterations in myelodysplastic syndrome (MDS) patients and occur in a mutually exclusive manner. The basis for the mutual exclusivity of these mutations and how they contribute to MDS is not well understood. Here we report that although different spliceosome gene mutations impart distinct effects on splicing, they are negatively selected for when co-expressed due to aberrant splicing and downregulation of regulators of hematopoietic stem cell survival and quiescence. In addition to this synthetic lethal interaction, mutations in the splicing factors SF3B1 and SRSF2 share convergent effects on aberrant splicing of mRNAs that promote nuclear factor κB signaling. These data identify shared consequences of splicing-factor mutations and the basis for their mutual exclusivity.

    View details for DOI 10.1016/j.ccell.2018.07.003

    View details for Web of Science ID 000441424600007

    View details for PubMedID 30107174

    View details for PubMedCentralID PMC6373472

  • MRD Testing in Multiple Myeloma: The Main Future Driver for Modern Tailored Treatment SEMINARS IN HEMATOLOGY Landgren, O., Lu, S. X., Hultcrantz, M. 2018; 55 (1): 44-50

    Abstract

    The past decade, several highly efficacious drugs have been approved for the treatment of multiple myeloma. Many of these newer drugs are less toxic than older chemotherapy drugs. Using modern combination therapy in newly diagnosed multiple myeloma patients, high proportions of newly diagnosed multiple myeloma patients obtain minimal residual disease (MRD) negativity and MRD testing has rapidly become an integral part of clinical trials focusing on patients in this setting. Only recently, MRD negativity was reported in clinical trials focusing on older newly diagnosed multiple myeloma patients (ie, nontransplant candidates), as well as studies focusing on patients with relapsed or refractory multiple myeloma. In the past, deeper responses were rarely seen in these patient categories due to inferior therapies and lack of MRD assays. The reason for the rapidly increased interest in MRD testing in all types of clinical trials is the fact that MRD negativity is closely correlated with longer progression-free survival which has been documented in recent meta-analyses. Consequently, MRD negativity has the potential to soon become a regulatory surrogate end-point for drug approval. This review dissects and discusses current data on MRD in multiple myeloma, it outlines new hypotheses, which can be tested in future clinical studies, and it discusses opportunities and future avenues for translational research. The goal of this article is to stimulate critical analysis of our current treatment landscape and development of future translational research involving MRD testing.

    View details for DOI 10.1053/j.seminhematol.2018.03.001

    View details for Web of Science ID 000433400100009

    View details for PubMedID 29759154

  • Genomic analysis of hairy cell leukemia identifies novel recurrent genetic alterations BLOOD Durham, B. H., Getta, B., Dietrich, S., Taylor, J., Won, H., Bogenberger, J. M., Scott, S., Kim, E., Chung, Y., Chung, S. S., Huellein, J., Walther, T., Wang, L., Lu, S. X., Oakes, C. C., Tibes, R., Haferlach, T., Taylor, B. S., Tallman, M. S., Berger, M. F., Park, J. H., Zenz, T., Abdel-Wahab, O. 2017; 130 (14): 1644-1648

    Abstract

    Classical hairy cell leukemia (cHCL) is characterized by a near 100% frequency of the BRAFV600E mutation, whereas ∼30% of variant HCLs (vHCLs) have MAP2K1 mutations. However, recurrent genetic alterations cooperating with BRAFV600E or MAP2K1 mutations in HCL, as well as those in MAP2K1 wild-type vHCL, are not well defined. We therefore performed deep targeted mutational and copy number analysis of cHCL (n = 53) and vHCL (n = 8). The most common genetic alteration in cHCL apart from BRAFV600E was heterozygous loss of chromosome 7q, the minimally deleted region of which targeted wild-type BRAF, subdividing cHCL into those hemizygous versus heterozygous for the BRAFV600E mutation. In addition to CDKN1B mutations in cHCL, recurrent inactivating mutations in KMT2C (MLL3) were identified in 15% and 25% of cHCLs and vHCLs, respectively. Moreover, 13% of vHCLs harbored predicted activating mutations in CCND3 A change-of-function mutation in the splicing factor U2AF1 was also present in 13% of vHCLs. Genomic analysis of de novo vemurafenib-resistant cHCL identified a novel gain-of-function mutation in IRS1 and losses of NF1 and NF2, each of which contributed to resistance. These data provide further insight into the genetic bases of cHCL and vHCL and mechanisms of RAF inhibitor resistance encountered clinically.

    View details for DOI 10.1182/blood-2017-01765107

    View details for Web of Science ID 000412274200007

    View details for PubMedID 28801450

    View details for PubMedCentralID PMC5630011

  • Robust patient-derived xenografts of MDS/MPN overlap syndromes capture the unique characteristics of CMML and JMML BLOOD Yoshimi, A., Balasis, M. E., Vedder, A., Feldman, K., Ma, Y., Zhang, H., Lee, S., Letson, C., Niyongere, S., Lu, S. X., Ball, M., Taylor, J., Zhang, Q., Zhao, Y., Youssef, S., Chung, Y., Zhang, X., Durham, B. H., Yang, W., List, A. F., Loh, M. L., Klimek, V., Berger, M. F., Stieglitz, E., Padron, E., Abdel-Wahab, O. 2017; 130 (4): 397-407

    Abstract

    Chronic myelomonocytic leukemia (CMML) and juvenile myelomonocytic leukemia (JMML) are myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) overlap disorders characterized by monocytosis, myelodysplasia, and a characteristic hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF). Currently, there are no available disease-modifying therapies for CMML, nor are there preclinical models that fully recapitulate the unique features of CMML. Through use of immunocompromised mice with transgenic expression of human GM-CSF, interleukin-3, and stem cell factor in a NOD/SCID-IL2Rγnull background (NSGS mice), we demonstrate remarkable engraftment of CMML and JMML providing the first examples of serially transplantable and genetically accurate models of CMML. Xenotransplantation of CD34+ cells (n = 8 patients) or unfractionated bone marrow (BM) or peripheral blood mononuclear cells (n = 10) resulted in robust engraftment of CMML in BM, spleen, liver, and lung of recipients (n = 82 total mice). Engrafted cells were myeloid-restricted and matched the immunophenotype, morphology, and genetic mutations of the corresponding patient. Similar levels of engraftment were seen upon serial transplantation of human CD34+ cells in secondary NSGS recipients (2/5 patients, 6/11 mice), demonstrating the durability of CMML grafts and functionally validating CD34+ cells as harboring the disease-initiating compartment in vivo. Successful engraftments of JMML primary samples were also achieved in all NSGS recipients (n = 4 patients, n = 12 mice). Engraftment of CMML and JMML resulted in overt phenotypic abnormalities and lethality in recipients, which facilitated evaluation of the JAK2/FLT3 inhibitor pacritinib in vivo. These data reveal that NSGS mice support the development of CMML and JMML disease-initiating and mature leukemic cells in vivo, allowing creation of genetically accurate preclinical models of these disorders.

    View details for DOI 10.1182/blood-2017-01-763219

    View details for Web of Science ID 000406492000008

    View details for PubMedID 28576879

    View details for PubMedCentralID PMC5533204

  • Splicing factor SF3B1(K700E) mutant dysregulates erythroid differentiation via aberrant alternative splicing of transcription factor TAL1 PLOS ONE Jin, S., Su, H., Ngoc-Tung Tran, Song, J., Lu, S. S., Li, Y., Huang, S., Abdel-Wahab, O., Liu, Y., Zhao, X. 2017; 12 (5): e0175523

    Abstract

    More than 60% of myeloid dysplasia syndrome (MDS) contains mutations in genes encoding for splicing factors such as SF3B1, U2AF, SRSF2 and ZRSR2. Mutations in SF3B1 are associated with 80% cases of refractory anemia with ring sideroblast (RARS), a subtype of MDS. SF3B1K700E is the most frequently mutated site among mutations on SF3B1. Yet the molecular mechanisms on how mutations of splicing factors lead to defective erythropoiesis are not clear. SF3B1K700E mutant binds to an RNA binding protein, RBM15, stronger than the wild type SF3B1 protein in co-immunoprecipitation assays. In addition, K700E mutant alters the RNA splicing of transcription factors TAL1 and GATA1. Via alternative RNA splicing, a novel short TAL1 transcript variant (TAL1s) is generated. Enhanced interaction between SF3B1 and RBM15 promotes the production of full-length TAL1 (TAL1fl) mRNA, while reduction of RBM15 protein level via PRMT1-mediated degradation pathway changes TAL1s/TAL1fl ratio in favor of TAL1s. TAL1s contains the helix-loop-helix DNA binding domain but not the N terminal region upstream of the DNA binding domain. The TAL1s protein loses its interaction with ETO2, which represses early erythropoiesis. In this vein, overexpression of TAL1s stimulates the transcription of β-hemoglobin in human leukemia K562 cells and promotes erythroid differentiation of human cord blood CD34+ cells cultured in erythropoietin-containing medium. Therefore, mutations of SF3B1 may block erythropoiesis via dysregulation of alternative RNA splicing of transcription factor TAL1, and targeting PRMT1 may alleviate the anemic symptoms in MDS patients.

    View details for DOI 10.1371/journal.pone.0175523

    View details for Web of Science ID 000401672400001

    View details for PubMedID 28545085

    View details for PubMedCentralID PMC5436638

  • Genetic drivers of vulnerability and resistance in relapsed acute lymphoblastic leukemia PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lu, S. X., Abdel-Wahab, O. 2016; 113 (40): 11071-11073

    View details for DOI 10.1073/pnas.1613836113

    View details for Web of Science ID 000384528900033

    View details for PubMedID 27663730

    View details for PubMedCentralID PMC5056046

  • Ceacam1 Separates Graft-versus-Host-Disease from Graft-versus-Tumor Activity after Experimental Allogeneic Bone Marrow Transplantation PLOS ONE Lu, S. X., Kappel, L. W., Charbonneau-Allard, A., Atallah, R., Holland, A. M., Turbide, C., Hubbard, V. M., Rotolo, J. A., Smith, M., Suh, D., King, C., Rao, U. K., Yim, N., Bautista, J. L., Jenq, R. R., Penack, O., Na, I., Liu, C., Murphy, G., Alpdogan, O., Blumberg, R. S., Macian, F., Holmes, K. V., Beauchemin, N., van den Brink, M. M. 2011; 6 (7): e21611

    Abstract

    Allogeneic bone marrow transplantation (allo-BMT) is a potentially curative therapy for a variety of hematologic diseases, but benefits, including graft-versus-tumor (GVT) activity are limited by graft-versus-host-disease (GVHD). Carcinoembryonic antigen related cell adhesion molecule 1 (Ceacam1) is a transmembrane glycoprotein found on epithelium, T cells, and many tumors. It regulates a variety of physiologic and pathological processes such as tumor biology, leukocyte activation, and energy homeostasis. Previous studies suggest that Ceacam1 negatively regulates inflammation in inflammatory bowel disease models.We studied Ceacam1 as a regulator of GVHD and GVT after allogeneic bone marrow transplantation (allo-BMT) in mouse models. In vivo, Ceacam1(-/-) T cells caused increased GVHD mortality and GVHD of the colon, and greater numbers of donor T cells were positive for activation markers (CD25(hi), CD62L(lo)). Additionally, Ceacam1(-/-) CD8 T cells had greater expression of the gut-trafficking integrin α(4)β(7), though both CD4 and CD8 T cells were found increased numbers in the gut post-transplant. Ceacam1(-/-) recipients also experienced increased GVHD mortality and GVHD of the colon, and alloreactive T cells displayed increased activation. Additionally, Ceacam1(-/-) mice had increased mortality and decreased numbers of regenerating small intestinal crypts upon radiation exposure. Conversely, Ceacam1-overexpressing T cells caused attenuated target-organ and systemic GVHD, which correlated with decreased donor T cell numbers in target tissues, and mortality. Finally, graft-versus-tumor survival in a Ceacam1(+) lymphoma model was improved in animals receiving Ceacam1(-/-) vs. control T cells.We conclude that Ceacam1 regulates T cell activation, GVHD target organ damage, and numbers of donor T cells in lymphoid organs and GVHD target tissues. In recipients of allo-BMT, Ceacam1 may also regulate tissue radiosensitivity. Because of its expression on both the donor graft and host tissues, this suggests that targeting Ceacam1 may represent a potent strategy for the regulation of GVHD and GVT after allogeneic transplantation.

    View details for DOI 10.1371/journal.pone.0021611

    View details for Web of Science ID 000292632000017

    View details for PubMedID 21760897

    View details for PubMedCentralID PMC3130781

  • Absence of P-Selectin in Recipients of Allogeneic Bone Marrow Transplantation Ameliorates Experimental Graft-versus-Host Disease JOURNAL OF IMMUNOLOGY Lu, S. X., Holland, A. M., Na, I., Terwey, T. H., Alpdogan, O., Bautista, J. L., Smith, O. M., Suh, D., King, C., Kochman, A., Hubbard, V. M., Rao, U. K., Yim, N., Liu, C., Laga, A. C., Murphy, G., Jenq, R. R., Zakrzewski, J. L., Penack, O., Dykstra, L., Bampoe, K., Perez, L., Furie, B., Furie, B., van den Brink, M. M. 2010; 185 (3): 1912-1919

    Abstract

    Alloreactive T cells are crucial for graft-versus-host disease (GVHD) pathophysiology, and modulating their trafficking patterns has been efficacious in ameliorating experimental disease. We report in this paper that P-selectin, a glycoprotein found on resting and inflamed endothelium, is important for donor alloreactive T cells trafficking into GVHD target organs, such as the intestines and skin. Compared with wild-type (WT) recipients of allogeneic bone marrow transplantation, P-selectin(-/-) recipients exhibit decreased GVHD mortality and decreased GVHD of the skin, liver, and small bowels. This was associated with diminished infiltration of alloactivated T cells into the Peyer's patches and small bowels, coupled with increased numbers of donor T cells in the spleen and secondary lymphoid organs (SLOs). Surprisingly, however, donor T cells deficient for P-selectin glycoprotein ligand 1, the most well described P-selectin ligand, mediated GVHD similar to WT T cells and accumulated in SLO and target organs in similar numbers as WT T cells. This suggests that P-selectin may be required for trafficking into inflamed tissues but not SLO and that donor T cells may use multiple P-selectin ligands apart from P-selectin glycoprotein ligand 1 to interact with P-selectin and traffic into inflamed tissues during GVHD. We conclude that targeting P-selectin may be a viable strategy for GVHD prophylaxis or treatment.

    View details for DOI 10.4049/jimmunol.0903148

    View details for Web of Science ID 000280177400069

    View details for PubMedID 20622117

    View details for PubMedCentralID PMC3752704

  • Inhibition of Neovascularization to Simultaneously Ameliorate Graft-vs-Host Disease and Decrease Tumor Growth JNCI-JOURNAL OF THE NATIONAL CANCER INSTITUTE Penack, O., Henke, E., Suh, D., King, C. G., Smith, O. M., Na, I., Holland, A. M., Ghosh, A., Lu, S. X., Jenq, R. R., Liu, C., Murphy, G. F., Lu, T. T., May, C., Scheinberg, D. A., Gao, D., Mittal, V., Heller, G., Benezra, R., van den Brink, M. M. 2010; 102 (12): 894-908

    Abstract

    BACKGROUND Blood vessels are formed either by sprouting of resident tissue endothelial cells (angiogenesis) or by recruitment of bone marrow (BM)-derived circulating endothelial progenitor cells (EPCs, vasculogenesis). Neovascularization has been implicated in tumor growth and inflammation, but its roles in graft-vs-host disease (GVHD) and in tumors after allogeneic BM transplantation (allo-BMT) were not known. METHODS We analyzed neovascularization, the contribution of endothelial cells and EPCs, and the ability of anti-vascular endothelial-cadherin antibody, E4G10, to inhibit neovascularization in mice with GVHD after allo-BMT using immunofluorescence microscopy and flow cytometry. We examined survival and clinical and histopathologic GVHD in mice (n = 10-25 per group) in which GVHD was treated with the E4G10 antibody using immunohistochemistry, flow cytometry, and cytokine immunoassay. We also assessed survival, the contribution of green fluorescent protein-marked EPCs to the tumor vasculature, and the ability of E4G10 to inhibit tumor growth in tumor-bearing mice (n = 20-33 per group) after allo-BMT using histopathology and bioluminescence imaging. All statistical tests were two-sided. RESULTS We found increased neovascularization mediated by vasculogenesis, as opposed to angiogenesis, in GVHD target tissues, such as liver and intestines. Administration of E4G10 inhibited neovascularization by donor BM-derived cells without affecting host vascularization, inhibited both GVHD and tumor growth, and increased survival (at 60 days post-BMT and tumor challenge with A20 lymphoma, the probability of survival was 0.29 for control antibody-treated allo-BMT recipients vs 0.7 for E4G10-treated allo-BMT recipients, 95% confidence interval = 0.180 to 0.640, P < .001). CONCLUSIONS Therapeutic targeting of neovascularization in allo-BMT recipients is a novel strategy to simultaneously ameliorate GVHD and inhibit posttransplant tumor growth, providing a new approach to improve the overall outcome of allogeneic hematopoietic stem cell transplantation.

    View details for DOI 10.1093/jnci/djq172

    View details for Web of Science ID 000279925200010

    View details for PubMedID 20463307

    View details for PubMedCentralID PMC2886094

  • Relapse after Allogeneic Hematopoietic Cell Therapy BIOLOGY OF BLOOD AND MARROW TRANSPLANTATION van den Brink, M. M., Porter, D. L., Giralt, S., Lu, S. X., Jenq, R. R., Hanash, A., Bishop, M. R. 2010; 16 (1): S138-S145

    Abstract

    Disease relapse remains a major cause of mortality following allogeneic hematopoietic cell transplantation (HCT). Over the past decade, our understanding of the biology underlying the graft-versus-tumor/leukemia (GVT) effect has increased greatly; however, several other factors affect the occurrence and outcome of relapse, including conditioning regimen, type of allograft, and the histology, status, and sensitivity to chemotherapy of the disease being treated. The mainstay of relapse treatment is donor lymphocyte infusion (DLI), but the efficacy of DLI is quite variable depending on disease histology and state. As such, there is a significant need for novel therapies and strategies for relapse following allogeneic HCT, particularly in patients for whom DLI is not an option. The National Cancer Institute is sponsoring an international workshop to address issues and research questions relative to the biology, natural history, prevention, and treatment of relapse following allogeneic HCT.

    View details for DOI 10.1016/j.bbmt.2009.10.023

    View details for Web of Science ID 000540636300023

    View details for PubMedID 19857588

    View details for PubMedCentralID PMC3637945

  • The cytolytic molecules Fas ligand and TRAIL are required for murine thymic graft-versus-host disease JOURNAL OF CLINICAL INVESTIGATION Na, I., Lu, S. X., Yim, N. L., Goldberg, G. L., Tsai, J., Rao, U., Smith, O. M., King, C. G., Suh, D., Hirschhorn-Cymerman, D., Palomba, L., Penack, O., Holland, A. M., Jenq, R. R., Ghosh, A., Tran, H., Merghoub, T., Liu, C., Sempowski, G. D., Ventevogel, M., Beauchemin, N., van den Brink, M. R. 2010; 120 (1): 343-356

    Abstract

    Thymic graft-versus-host disease (tGVHD) can contribute to profound T cell deficiency and repertoire restriction after allogeneic BM transplantation (allo-BMT). However, the cellular mechanisms of tGVHD and interactions between donor alloreactive T cells and thymic tissues remain poorly defined. Using clinically relevant murine allo-BMT models, we show here that even minimal numbers of donor alloreactive T cells, which caused mild nonlethal systemic graft-versus-host disease, were sufficient to damage the thymus, delay T lineage reconstitution, and compromise donor peripheral T cell function. Furthermore, to mediate tGVHD, donor alloreactive T cells required trafficking molecules, including CCR9, L selectin, P selectin glycoprotein ligand-1, the integrin subunits alphaE and beta7, CCR2, and CXCR3, and costimulatory/inhibitory molecules, including Ox40 and carcinoembryonic antigen-associated cell adhesion molecule 1. We found that radiation in BMT conditioning regimens upregulated expression of the death receptors Fas and death receptor 5 (DR5) on thymic stromal cells (especially epithelium), while decreasing expression of the antiapoptotic regulator cellular caspase-8-like inhibitory protein. Donor alloreactive T cells used the cognate proteins FasL and TNF-related apoptosis-inducing ligand (TRAIL) (but not TNF or perforin) to mediate tGVHD, thereby damaging thymic stromal cells, cytoarchitecture, and function. Strategies that interfere with Fas/FasL and TRAIL/DR5 interactions may therefore represent a means to attenuate tGVHD and improve T cell reconstitution in allo-BMT recipients.

    View details for DOI 10.1172/JCI39395

    View details for Web of Science ID 000273495700036

    View details for PubMedID 19955659

    View details for PubMedCentralID PMC2798682

  • NOD2 Regulates Hematopoietic Cell Function During Graft-Versus-Host Disease. Penack, O., Smith, O. M., Yim, N., Rao, U., Ghosh, A., Murphy, G. F., Holland, A. M., Jenq, R. R., Lu, S. X., Na, I., van den Brink, M. M. AMER SOC HEMATOLOGY. 2009: 967-968
  • TRAIL/DR5 Interactions Are Important for Thymic Damage After Allogeneic Bone Marrow Transplantation Na, I., Lu, S. X., Yim, N. L., Goldberg, G. L., Tsai, J., Rao, U. K., Smith, O. M., King, C., Suh, D., Hirschhorn-Cymerman, D., Palomba, M., Penack, O., Holland, A. M., Jenq, R. R., Ghosh, A., Tran, H., Merghoub, T., Liu, C., Sempowski, G., Ventevogel, M., Beauchemin, N., Furie, B., van den Brink, M. M. AMER SOC HEMATOLOGY. 2009: 101
  • Cytolytic T cells induce ceramide-rich platforms in target cell membranes to initiate graft-versus-host disease BLOOD Rotolo, J. A., Stancevic, B., Lu, S. X., Zhang, J., Suh, D., King, C. G., Kappel, L. W., Murphy, G. F., Liu, C., Fuks, Z., van den Brink, M. R., Kolesnick, R. 2009; 114 (17): 3693-3706

    Abstract

    Alloreactive donor cytolytic T lymphocytes play a critical role in pathophysiology of acute graft-versus-host disease (GVHD). As GVHD progression involves tumor necrosis factor superfamily receptor activation, and as apoptotic signaling for some tumor necrosis factor superfamily receptors might involve acid sphingomyelinase (ASMase)-mediated ceramide generation, we hypothesized that ASMase deletion would ameliorate GVHD. Using clinically relevant mouse models of acute GVHD in which allogeneic bone marrow and T cells were transplanted into asmase+/+ and asmase(-/-) hosts, we identify host ASMase as critical for full-blown GVHD. Lack of host ASMase reduced the acute inflammatory phase of GVHD, attenuating cytokine storm, CD8+ T-cell proliferation/activation, and apoptosis of relevant graft-versus-host target cells (hepatocytes, intestinal, and skin cells). Organ injury was diminished in asmase(-/-) hosts, and morbidity and mortality improved at 90 days after transplantation. Resistance to cytolytic T lymphocyte-induced apoptosis was found at the target cell membrane if hepatocytes lack ASMase, as hepatocyte apoptosis required target cell ceramide generation for formation of ceramide-rich macrodomains, sites concentrating proapoptotic Fas. These studies indicate a requirement for target cell ASMase in evolution of GVHD in liver, small intestines, and skin and provide potential new targets for disease management.

    View details for DOI 10.1182/blood-2008-11-191148

    View details for Web of Science ID 000271024500026

    View details for PubMedID 19666872

    View details for PubMedCentralID PMC2766684

  • NOD2 regulates hematopoietic cell function during graft-versus-host disease JOURNAL OF EXPERIMENTAL MEDICINE Penack, O., Smith, O. M., Cunningham-Bussel, A., Liu, X., Rao, U., Yim, N., Na, I., Holland, A. M., Ghosh, A., Lu, S. X., Jenq, R. R., Liu, C., Murphy, G. F., Brandl, K., van den Brink, M. M. 2009; 206 (10): 2101-2110

    Abstract

    Nucleotide-binding oligomerization domain 2 (NOD2) polymorphisms are independent risk factors for Crohn's disease and graft-versus-host disease (GVHD). In Crohn's disease, the proinflammatory state resulting from NOD2 mutations have been associated with a loss of antibacterial function of enterocytes such as paneth cells. NOD2 has not been studied in experimental allogeneic bone marrow transplantation (allo-BMT). Using chimeric recipients with NOD2(-/-) hematopoietic cells, we demonstrate that NOD2 deficiency in host hematopoietic cells exacerbates GVHD. We found that proliferation and activation of donor T cells was enhanced in NOD-deficient allo-BMT recipients, suggesting that NOD2 plays a role in the regulation of host antigen-presenting cells (APCs). Next, we used bone marrow chimeras in an experimental colitis model and observed again that NOD2 deficiency in the hematopoietic cells results in increased intestinal inflammation. We conclude that NOD2 regulates the development of GVHD through its inhibitory effect on host APC function.

    View details for DOI 10.1084/jem.20090623

    View details for Web of Science ID 000270269300006

    View details for PubMedID 19737867

    View details for PubMedCentralID PMC2757869

  • The T Cell Cytolytic Molecules FasL and TRAIL are Required for Thymic GVHD Lu, S. X., Na, I., Goldberg, G. L., King, C. G., Smith, M., Suh, D., Rao, U., Yim, N., Holland, A. M., Penack, O., Jenq, R. R., Ghosh, A., Houghton, A., van den Brink, M. M. AMER ASSOC IMMUNOLOGISTS. 2009
  • NOD2 deficiency in experimental GvHD Penack, O., Smith, O., Liu, X., Cunningham, A., Rao, U., Jim, N., Na, I., Holland, A., Ghosh, A., Lu, S., Jenq, R., Ubeda, C., Brandl, K., Pamer, E., Van den Brink, M. NATURE PUBLISHING GROUP. 2009: S51
  • Inhibition of vasculogenesis ameliorates GvHD and tumours after allo-BMT Penack, O., Henke, E., Suh, D., King, C., Smith, O., Na, I., Holland, A., Ghosh, A., Lu, S., Jenq, R., Liu, C., Murphy, G., Lu, T., Scheinberg, D., Gao, D. C., Mittal, V., Benezra, R., Van den Brink, M. NATURE PUBLISHING GROUP. 2009: S20-S21
  • Keratinocyte growth factor enhances DNA plasmid tumor vaccine responses after murine allogeneic bone marrow transplantation BLOOD Jenq, R. R., King, C. G., Volk, C., Suh, D., Smith, O. M., Rao, U. K., Yim, N. L., Holland, A. M., Lu, S. X., Zakrzewski, J. L., Goldberg, G. L., Diab, A., Alpdogan, O., Penack, O., Na, I., Kappel, L. W., Wolchok, J. D., Houghton, A. N., Perales, M., van den Brink, M. M. 2009; 113 (7): 1574-1580

    Abstract

    Keratinocyte growth factor (KGF), which is given exogenously to allogeneic bone marrow transplantation (allo-BMT) recipients, supports thymic epithelial cells and increases thymic output of naive T cells. Here, we demonstrate that this improved T-cell reconstitution leads to enhanced responses to DNA plasmid tumor vaccination. Tumor-bearing mice treated with KGF and DNA vaccination have improved long-term survival and decreased tumor burden after allo-BMT. When assayed before vaccination, KGF-treated allo-BMT recipients have increased numbers of peripheral T cells, including CD8(+) T cells with vaccine-recognition potential. In response to vaccination, KGF-treated allo-BMT recipients, compared with control subjects, generate increased numbers of tumor-specific CD8(+) cells, as well as increased numbers of CD8(+) cells producing interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). We also found unanticipated benefits to antitumor immunity with the administration of KGF. KGF-treated allo-BMT recipients have an improved ratio of T effector cells to regulatory T cells, a larger fraction of effector cells that display a central memory phenotype, and effector cells that are derived from a broader T-cell-receptor repertoire. In conclusion, our data suggest that KGF can function as a potent vaccine adjuvant after allo-BMT through its effects on posttransplantation T-cell reconstitution.

    View details for DOI 10.1182/blood-2008-05-155697

    View details for Web of Science ID 000263316400025

    View details for PubMedID 19011222

    View details for PubMedCentralID PMC2644085

  • CEACAM1 REGULATES EXPERIMENTAL GRAFT-VERSUS-HOST-DISEASE Lu, S. X., Willis, L., Charbonneau-Allard, A. M., Atallah, R., Holland, A. M., Turbide, C., Hubbard, V. M., Rotolo, J. A., Smith, O. M., Suh, D., King, C., Rao, U. K., Yim, N., Kochman, A., Bautista, J. L., Jenq, R. R., Zakrzewski, J. L., Tran, H., Penack, O., Na, I. K., Chow, M., Lin, J., Cabrera-Perez, J., Liu, C., Murphy, G., Alpdogan, O., Blumberg, R. S., Macian, F., Holmes, K., Beauchemin, N., van den Brink, M. M. ELSEVIER SCIENCE INC. 2009: 13
  • DEPLETION OF VASCULAR ENDOTHELIAL PROGENITOR CELLS SIMULTANEOUSLY AMELIORATES GVHD AND INHIBITS TUMOR GROWTH Penack, O., Henke, E., Suh, D., King, C., Smith, M., Na, I., Holland, A., Ghosh, A., Lu, S., Feng, R., Liu, C., May, C., Murphy, G., Lu, T., Gao, D., Mittal, V., Benezra, R., van den Brink, M. ELSEVIER SCIENCE INC. 2009: 1
  • STAT-3 and ERK 1/2 phosphorylation are critical for T-cell alloactivation and graft-versus-host disease BLOOD Lu, S. X., Alpdogan, O., Lin, J., Balderas, R., Campos-Gonzalez, R., Wang, X., Gao, G., Suh, D., King, C., Chow, M., Smith, O. M., Hubbard, V. M., Bautista, J. L., Cabrera-Perez, J., Zakrzewski, J. L., Kochman, A. A., Chow, A., Altan-Bonnet, G., van den Brink, M. M. 2008; 112 (13): 5254-5258

    Abstract

    Graft-versus-host disease (GVHD) is a serious complication of allogeneic bone marrow transplantation, and donor T cells are indispensable for GVHD. Current therapies have limited efficacy, selectivity, and high toxicities. We used a novel flow cytometry technique for the analysis of intracellular phosphorylation events in single cells in murine BMT models to identify and validate novel GVHD drug targets.(1-7) This method circumvents the requirement for large numbers of purified cells, unlike western blots. We defined a signaling profile for alloactivated T cells in vivo and identified the phosphorylation of ERK1/2 and STAT-3 as important events during T-cell (allo)activation in GVHD. We establish that interference with STAT-3 phosphorylation can inhibit T-cell activation and proliferation in vitro and GVHD in vivo. This suggests that phospho-specific flow cytometry is useful for the identification of promising drug targets, and ERK1/2 and STAT-3 phosphorylation in alloactivated T cells may be important for GVHD.

    View details for DOI 10.1182/blood-2008-03-147322

    View details for Web of Science ID 000261513400061

    View details for PubMedID 18838616

    View details for PubMedCentralID PMC2597618

  • Rapidly proliferating CD44(hi) peripheral T cells undergo apoptosis and delay posttransplantation T-cell reconstitution after allogeneic bone marrow transplantation BLOOD Alpdogan, S., Lu, S. X., Patel, N., McGoldrick, S., Suh, D., Budak-Alpdogan, T., Smith, O. M., Grubin, J., King, C., Goldberg, G. L., Hubbard, V. M., kochman, A. A., van den Brink, M. M. 2008; 112 (12): 4755-4764

    Abstract

    Delayed T-cell recovery is an important complication of allogeneic bone marrow transplantation (BMT). We demonstrate in murine models that donor BM-derived T cells display increased apoptosis in recipients of allogeneic BMT with or without GVHD. Although this apoptosis was associated with a loss of Bcl-2 and Bcl-X(L) expression, allogeneic recipients of donor BM deficient in Fas-, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)- or Bax-, or BM-overexpressing Bcl-2 or Akt showed no decrease in apoptosis of peripheral donor-derived T cells. CD44 expression was associated with an increased percentage of BM-derived apoptotic CD4(+) and CD8(+) T cells. Transplantation of RAG-2-eGFP-transgenic BM revealed that proliferating eGFP(lo)CD44(hi) donor BM-derived mature T cells were more likely to undergo to apoptosis than nondivided eGFP(hi)CD44(lo) recent thymic emigrants in the periphery. Finally, experiments using carboxyfluorescein succinimidyl ester-labeled T cells adoptively transferred into irradiated syngeneic hosts revealed that rapid spontaneous proliferation (as opposed to slow homeostatic proliferation) and acquisition of a CD44(hi) phenotype was associated with increased apoptosis in T cells. We conclude that apoptosis of newly generated donor-derived peripheral T cells after an allogeneic BMT contributes to delayed T-cell reconstitution and is associated with CD44 expression and rapid spontaneous proliferation by donor BM-derived T cells.

    View details for DOI 10.1182/blood-2008-02-142737

    View details for Web of Science ID 000261217000055

    View details for PubMedID 18815289

    View details for PubMedCentralID PMC2597141

  • Depletion of Vascular Endothelial Progenitor Cells Inhibits Inflammation Penack, O., Herike, E., Suh, D., King, C., Smith, M. O., Na, I., Holland, A., Ghosh, A., Lu, S. X., Jenq, R., Liu, C., Murphy, G. F., Lu, T., May, C., Scheinberg, D. A., Gao, D., Mittal, V., Benezra, R., Van Den Brink, M. AMER SOC HEMATOLOGY. 2008: 258-259
  • Signal transduction in chronic lymphocytic leukemia (CLL) cells by multiparameter flow-cytometry Palomba, M. L., Patel, J. P., Rao, U., Rastalsky, N., Lu, S. X., Houghton, A. N., Zelenetz, A. D., van den Brink, M. R. ELSEVIER. 2008: 192
  • Tumor immunotherapy across MHC barriers using allogeneic T-cell precursors NATURE BIOTECHNOLOGY Zakrzewski, J. L., Suh, D., Markley, J. C., Smith, O. M., King, C., Goldberg, G. L., Jenq, R., Holland, A. M., Grubin, J., Cabrera-Perez, J., Brentjens, R. J., Lu, S. X., Rizzuto, G., Sant'Angelo, D. B., Riviere, I., Sadelain, M., Heller, G., Zuniga-Pflucker, J., Lu, C., van den Brink, M. M. 2008; 26 (4): 453-461

    Abstract

    We present a strategy for adoptive immunotherapy using T-lineage committed lymphoid precursor cells generated by Notch1-based culture. We found that allogeneic T-cell precursors can be transferred to irradiated individuals irrespective of major histocompatibility complex (MHC) disparities and give rise to host-MHC restricted and host-tolerant functional allogeneic T cells, improving survival in irradiated recipients as well as enhancing anti-tumor responses. T-cell precursors transduced to express a chimeric receptor targeting hCD19 resulted in significant additional anti-tumor activity, demonstrating the feasibility of genetic engineering of these cells. We conclude that ex vivo generated MHC-disparate T-cell precursors from any donor can be used universally for 'off-the-shelf' immunotherapy, and can be further enhanced by genetic engineering for targeted immunotherapy.

    View details for DOI 10.1038/nbt1395

    View details for Web of Science ID 000254782500028

    View details for PubMedID 18376399

    View details for PubMedCentralID PMC2731996

  • Off-the-shelf tumor immunotherapy with genetically enhanced allogeneic T cell precursors Zakrzewski, J. L., Suh, D., Markley, J., King, C., Smith, O. M., Goldberg, G., Jenq, R., Holland, A. M., Lu, S., Sant'Angelo, D., Riviere, I., Sadelain, M., van den Brink, M. M. FEDERATION AMER SOC EXP BIOL. 2008
  • OFF-THE-SHELF TUMOR IMMUNOTHERAPY WITH GENETICALLY ENHANCED ALLOGENEIC T-CELL PRECURSORS Zakrzewski, J. L., Suh, D., Markley, J. C., Smith, O. M., King, C., Goldberg, G., Jenq, R., Holland, A. M., Grubin, J., Cabrera-Perez, J., Lu, S. X., Rizzuto, G., Sant'Angelo, D. B., Riviere, Sadelain, M., Zuniga-Pflucker, J. C., van den Brink, M. M. ELSEVIER SCIENCE INC. 2008: 126
  • Discovery and validation of STAT-3 and ERK1/2 phosphorylation as critical for the function of alloactivated T cells in acute graft-versus-host-disease via a novel technique for drug discovery Lu, S. X., Alpdogan, O., Campos, R., Wang, X., Gao, G., Chow, M., Lin, J., Suh, D., King, C., Hubbard, V. M., Bautista, J. L., Kochman, A. A., Zakrzewski, J. L., Smith, O., Grubin, J., Chow, A., Patel, N., van den Brink, M. M. AMER SOC HEMATOLOGY. 2007: 950A-951A
  • Targeted 'Off-the-Shelf' tumor immunotherapy using allogeneic T-Cell precursors Zakrzewski, J. L., Suh, D., Smith, O. M., King, C., Goldberg, G. L., Jenq, R., Holland, A. M., Grubin, J., Lu, S. X., Cabrera-Perez, J., Rizzuto, G., Sant'Angelo, D. B., van den Brink, M. M., Markley, J. C., Riviere, I., Sadelain, M., Zuniga-Pflucker, J. AMER SOC HEMATOLOGY. 2007: 178A
  • CEACAM-1 is involved in graft-versus-host-discase in murine allogeneic bone marrow transplantation models Lu, S. X., Willis, L., Smith, M., Suh, D., King, C., Bautista, J., Chow, M., Cabrera-Perez, J., Hubbard, V., Rotolo, J., Liu, C., Murphy, G., Alpdogan, O., Blumberg, R., Holmes, K., Turbide, C., Beauchemin, N., van den Brink, M. M. AMER SOC HEMATOLOGY. 2007: 28A
  • IFN-gamma and Fas ligand are required for graft-versus-tumor activity against renal cell carcinoma in the absence of lethal graft-versus-host disease JOURNAL OF IMMUNOLOGY Ramirez-Montagut, T., Chow, A., Kochman, A. A., Smith, O. M., Suh, D., Sindhi, H., Lu, S., Borsotti, C., Grubin, J., Patel, N., Terwey, T. H., Kim, T. D., Heller, G., Murphy, G. F., Liu, C., Alpdogan, O., van den Brink, M. M. 2007; 179 (3): 1669-1680

    Abstract

    To determine the mechanisms of graft-versus-tumor (GVT) activity in the absence of graft-versus-host disease (GVHD) against a solid tumor, we established two allogeneic bone marrow transplantation models with a murine renal cell carcinoma (RENCA). The addition of 0.3 x 10(6) donor CD8(+) T cells to the allograft increased the survival of tumor-bearing mice without causing GVHD. The analysis of CD8(+) T cells deficient in cytotoxic molecules demonstrated that anti-RENCA activity is dependent on IFN-gamma and Fas ligand (FasL), but does not require soluble or membrane-bound TNF-alpha, perforin, or TRAIL. Recipients of IFN-gamma(-/-) CD8(+) T cells are unable to reject RENCA compared with recipients of wild-type CD8(+) T cells and, importantly, neither group develops severe GVHD. IFN-gamma(-/-) CD8(+) T cells derived from transplanted mice are less able to kill RENCA cells in vitro, while pretreatment of RENCA cells with IFN-gamma enhances class I and FasL expression and rescues the lytic capacity of IFN-gamma(-/-) CD8(+) T cells. These results demonstrate that the addition of low numbers of selected donor CD8(+) T cells to the allograft can mediate GVT activity without lethal GVHD against murine renal cell carcinoma, and this GVT activity is dependent on IFN-gamma and FasL.

    View details for DOI 10.4049/jimmunol.179.3.1669

    View details for Web of Science ID 000248319700030

    View details for PubMedID 17641033

  • Absence of donor T-cell-derived soluble TNF decreases graft-versus-host disease without impairing graft-versus-tumor activity BLOOD Borsotti, C., Franklin, A. K., Lu, S. X., Kim, T. D., Smith, O. M., Suh, D., King, C. G., Chow, A., Liu, C., Alpdogan, O., van den Brink, M. M. 2007; 110 (2): 783-786

    Abstract

    Tumor necrosis factor (TNF) plays an important role in graft-versus-host disease (GVHD) and graft-versus-tumor (GVT) activity after allogeneic bone marrow transplantation (allo-BMT). TNF can be expressed in a membrane-bound form (memTNF) and as a soluble (solTNF) molecule after being cleaved by the TNF-alpha converting enzyme (TACE). To study the contribution of donor T-cell-derived memTNF versus solTNF in GVHD and GVT, we used mice containing a noncleavable allele in place of endogenous TNF (memTNF(Delta/Delta)) as donors in murine BMT models. Recipients of memTNF T cells developed significantly less GVHD than recipients of wild-type (wt) T cells. In contrast, GVT activity mediated by memTNF T cells remained intact, and alloreactive memTNF T cells showed no defects in proliferation, activation, and cytotoxicity. These data suggest that suppressing the secretion of solTNF by donor T cells significantly decreases GVHD without impairing GVT activity.

    View details for DOI 10.1182/blood-2006-10-054510

    View details for Web of Science ID 000248112400043

    View details for PubMedID 17395784

    View details for PubMedCentralID PMC1924485

  • Peripheral T cell apoptosis is associated with homeostatic T cell proliferation and plays a significant role in post-transplant immune deficiency. Alpdogan, O., Patel, N., Suh, D., Grubin, J., McGoldrick, S., Smith, O. M., Lu, S., King, C., Kochman, A., Hubbard, V., van den Brink, M. M. AMER SOC HEMATOLOGY. 2006: 25A
  • Adoptive transfer of T-cell precursors enhances T-cell reconstitution after allogeneic hematopoietic stem cell transplantation NATURE MEDICINE Zakrzewski, J. L., Kochman, A. A., Lu, S. X., Terwey, T. H., Kim, T. D., Hubbard, V. M., Muriglan, S. J., Suh, D., Smith, O. M., Grubin, J., Patel, N., Chow, A., Cabrera-Perez, J., Radhakrishnan, R., Diab, A., Perales, M., Rizzuto, G., Menet, E., Pamer, E. G., Heller, G., Zuniga-Pflucker, J., Alpdogan, O., van den Brink, M. M. 2006; 12 (9): 1039-1047

    Abstract

    Immunoincompetence after allogeneic hematopoietic stem cell transplantation (HSCT) affects in particular the T-cell lineage and is associated with an increased risk for infections, graft failure and malignant relapse. To generate large numbers of T-cell precursors for adoptive therapy, we cultured mouse hematopoietic stem cells (HSCs) in vitro on OP9 mouse stromal cells expressing the Notch-1 ligand Delta-like-1 (OP9-DL1). We infused these cells, together with T-cell-depleted mouse bone marrow or purified HSCs, into lethally irradiated allogeneic recipients and determined their effect on T-cell reconstitution after transplantation. Recipients of OP9-DL1-derived T-cell precursors showed increased thymic cellularity and substantially improved donor T-cell chimerism (versus recipients of bone marrow or HSCs only). OP9-DL1-derived T-cell precursors gave rise to host-tolerant CD4+ and CD8+ populations with normal T-cell antigen receptor repertoires, cytokine secretion and proliferative responses to antigen. Administration of OP9-DL1-derived T-cell precursors increased resistance to infection with Listeria monocytogenes and mediated significant graft-versus-tumor (GVT) activity but not graft-versus-host disease (GVHD). We conclude that the adoptive transfer of OP9-DL1-derived T-cell precursors markedly enhances T-cell reconstitution after transplantation, resulting in GVT activity without GVHD.

    View details for DOI 10.1038/nm1463

    View details for Web of Science ID 000240373900028

    View details for PubMedID 16936725

  • Glucocorticoid-induced TNF receptor family related gene activation overcomes tolerance/ignorance to melanoma differentiation antigens and enhances antitumor immunity JOURNAL OF IMMUNOLOGY Ramirez-Montagut, T., Chow, A., Hirschhorn-Cymerman, D., Terwey, T. H., Kochman, A. A., Lu, S., Miles, R. C., Sakaguchi, S., Houghton, A. N., van den Brink, M. M. 2006; 176 (11): 6434-6442

    Abstract

    Glucocorticoid-induced TNF receptor family related protein (GITR) is present on many different cell types. Previous studies have shown that in vivo administration of an anti-GITR agonist mAb (DTA-1) inhibits regulatory T cells (Treg)-dependent suppression and enhances T cell responses. In this study, we show that administration of DTA-1 induces >85% tumor rejection in mice challenged with B16 melanoma. Rejection requires CD4+, CD8+, and NK1.1+ cells and is dependent on IFN-gamma and Fas ligand and independent of perforin. Depletion of Treg via anti-CD25 treatment does not induce B16 rejection, whereas 100% of the mice depleted of CD25+ cells and treated with DTA-1 reject tumors, indicating a predominant role of GITR on effector T cell costimulation rather than on Treg modulation. T cells isolated from DTA-1-treated mice challenged with B16 are specific against B16 and several melanoma differentiation Ags. These mice develop memory against B16, and a small proportion of them develop mild hypopigmentation. Consistent with previous studies showing that GITR stimulation increases Treg proliferation in vitro, we found in our model that GITR stimulation expanded the absolute number of FoxP3+ cells in vivo. Thus, we conclude that overall, GITR stimulation overcomes self-tolerance/ignorance and enhances T cell-mediated antitumor activity with minimal autoimmunity.

    View details for DOI 10.4049/jimmunol.176.11.6434

    View details for Web of Science ID 000237754200007

    View details for PubMedID 16709800

  • Keratinocyte growth factor (KGF) is required for postnatal thymic regeneration BLOOD Alpdogan, O., Hubbard, V. M., Smith, O. M., Patel, N., Lu, S., Goldberg, G. L., Gray, D. H., Feinman, J., Kochman, A. A., Eng, J. M., Suh, D., Muriglan, S. J., Boyd, R. L., van den Brink, M. R. 2006; 107 (6): 2453-2460

    Abstract

    Keratinocyte growth factor (KGF) is a member of the fibroblast growth factor family that mediates epithelial cell proliferation and differentiation in a variety of tissues, including the thymus. We studied the role of KGF in T-cell development with KGF-/- mice and demonstrated that thymic cellularity and the distribution of thymocyte subsets among KGF-/-, wildtype (WT), and KGF+/- mice were similar. However, KGF-/- mice are more vulnerable to sublethal irradiation (450 cGy), and a significant decrease was found in thymic cellularity after irradiation. Defective thymopoiesis and peripheral T-cell reconstitution were found in KGF-/- recipients of syngeneic or allogeneic bone marrow transplant, but using KGF-/- mice as a donor did not affect T-cell development after transplantation. Despite causing an early developmental block in the thymus, administration of KGF to young and old mice enhanced thymopoiesis. Exogenous KGF also accelerated thymic recovery after irradiation, cyclophosphamide, and dexamethasone treatment. Finally, we found that administering KGF before bone marrow transplantation (BMT) resulted in enhanced thymopoiesis and peripheral T-cell numbers in middle-aged recipients of an allogeneic BM transplant. We conclude that KGF plays a critical role in postnatal thymic regeneration and may be useful in treating immune deficiency conditions.

    View details for DOI 10.1182/blood-2005-07-2831

    View details for Web of Science ID 000236014200039

    View details for PubMedID 16304055

    View details for PubMedCentralID PMC1895735

  • Absence of beta 7 integrin results in less graft-versus-host disease because of decreased homing of alloreactive T cells to intestine BLOOD Waldman, E., Lu, S. X., Hubbard, V. M., Kochman, A. A., Eng, J. M., Terwey, T. H., Muriglan, S. J., Kim, T. D., Heller, G., Murphy, G. F., Liu, C., Alpdogan, O., van den Brink, M. R. 2006; 107 (4): 1703-1711

    Abstract

    The alpha4beta7 integrin plays a central role in the homing of T cells to the gut. We hypothesized that absence of the beta7 subunit would result in a reduction of intestinal graft-versus-host disease (GVHD) and an improvement in overall GVHD morbidity and mortality in recipients of hematopoietic stem cell transplantation (HSCT). Analysis of alloreactive beta7-/- T cells showed intact activation, proliferation, cytokine production, and cytotoxicity. However, recipients of beta7-/- donor T cells in murine HSCT models experienced less GVHD morbidity and mortality than recipients of wild-type (WT) T cells, associated with a decrease in donor T-cell infiltration of the liver and intestine and with an overall significant decrease in hepatic and intestinal GVHD. In graft-versus-tumor (GVT) experiments, we demonstrated intact or even enhanced GVT activity of beta7-/- donor T cells. In conclusion, beta7-/- donor T cells caused less GVHD morbidity and mortality than WT donor T cells because of selectively decreased T-cell infiltration of the liver and intestines. Our data suggest that strategies to target the beta7 integrin have the clinical potential to alleviate or prevent GVHD while sparing or potentiating GVT activity.

    View details for DOI 10.1182/blood-2005-08-3445

    View details for Web of Science ID 000235296100067

    View details for PubMedID 16291587

    View details for PubMedCentralID PMC1895413

  • Peripheral T cell apoptosis affects immune reconstitution after allogeneic BMT Alpdogan, O., McGoldrick, S., Lu, S., Patel, N., Suh, D., Smith, O. M., van den Brink, M. R. CARDEN JENNINGS PUBL CO LTD. 2006: 81
  • Adoptive transfer of in vitro generated T cell precursors improves T cell reconstitution and mediates graft-versus-tumor activity without graft-versus-host disease in allogeneic hematopoietic stem cell transplantation recipients Zakrzewski, J. L., Kochman, A. A., Lu, S. X., Terwey, T., Kim, T. D., Hubbard, V. M., Muriglan, S. J., Suh, D., Cabrera-Perez, J., Heller, G., Zuniga-Pflucker, J. C., Alodogan, O., van den Brink, M. R. CARDEN JENNINGS PUBL CO LTD. 2006: 82
  • T-cell signaling in allogeneic bone marrow transplantation Lu, S. X., Chow, M. Y., Hubbard, V. M., Suh, D. Y., Kim, T. D., Kochman, A., Zakrzewski, J., Cabrera-Perez, J., Alpdogan, O., van den Brink, M. R. CARDEN JENNINGS PUBL CO LTD. 2006: 24
  • Signaling profiles of T cells during allogeneic hematopoietic stem cell transplantation. Lu, S. X., Hubbard, V. M., Chow, M. Y., Kim, T. D., Suh, D. Y., Kochman, A., Zakrzewski, J., Cabrera-Perez, J., Alpdogan, O., van den Brink, M. M. AMER SOC HEMATOLOGY. 2005: 367A-368A
  • Adoptive transfer of in vitro generated T cell precursors enhances donor T cell reconstitution and graft-versus-tumor activity in allogeneic hematopoietic stem cell transplantation recipients. Zakrzewski, J. L., Kochman, A. A., Lu, S. X., Terwey, T. H., Kim, T. D., Hubbard, V. M., Muriglan, S. J., Suh, D., Cabrera-Perez, J., Radhakrishnan, R., Heller, G., Zuniga-Pflucker, J. C., Alpdogan, O., van den Brink, M. R. AMER SOC HEMATOLOGY. 2005: 23A
  • CCR2 is required for CD8-induced graft-versus-host disease BLOOD Terwey, T. H., Kim, T. D., Kochman, A. A., Hubbard, V. M., Lu, S., Zakrzewski, J. L., Ramirez-Montagut, T., Eng, J. M., Muriglan, S. J., Heller, G., Murphy, G. F., Liu, C., Budak-Alpdogan, T., Alpdogan, O., van den Brink, M. R. 2005; 106 (9): 3322-3330

    Abstract

    Graft-versus-host disease (GVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (HSCT). Migration of donor-derived T cells into GVHD target organs plays a critical role in the development of GVHD and chemokines and their receptors are important molecules involved in this process. Here, we demonstrate in murine bone marrow transplantation models that the expression of the inflammatory CC chemokine receptor 2 (CCR2) on donor-derived CD8+ T cells is relevant for the control of CD8+ T-cell migration and development of GVHD. Recipients of CCR2-deficient (CCR2-/-) CD8+ T cells developed less damage of gut and liver than recipients of wild-type CD8+ T cells, which correlated with a reduction in overall GVHD morbidity and mortality. Assessment of donor CD8+ T-cell target organ infiltration revealed that CCR2-/- CD8+ T cells have an intrinsic migratory defect to the gut and liver. Other causes for the reduction in GVHD could be excluded, as alloreactive proliferation, activation, IFN-gamma production and cytotoxicity of CCR2-/- CD8+ T cells were intact. Interestingly, the graft-versus-tumor effect mediated by CCR2-/- CD8+ T cells was preserved, which suggests that interference with T-cell migration by blockade of CCR2 signaling can separate GVHD from GVT activity.

    View details for DOI 10.1182/blood-2005-05-1860

    View details for Web of Science ID 000232917400065

    View details for PubMedID 16037386

    View details for PubMedCentralID PMC1895329

  • Adoptive transfer of in vitro-generated T cell precursors enhances posttransplant donor T cell reconstitution in recipients of an allogeneic hematopoietic stem cell transplantation Zakrzewski, J. L., Kochman, A., Muriglan, S. J., Lu, S., Kim, T. D., Zuniga-Pflucker, J. C., Alpdogan, O., van den Brink, M. R. CARDEN JENNINGS PUBL CO LTD. 2005: 4-5