scispace - formally typeset
Search or ask a question

Showing papers by "Robert P. Hasserjian published in 2020"


Journal ArticleDOI
Elsa Bernard1, Yasuhito Nannya2, Robert P. Hasserjian3, Sean M. Devlin1, Heinz Tuechler, Juan S. Medina-Martinez1, Tetsuichi Yoshizato2, Yusuke Shiozawa2, Ryunosuke Saiki2, Luca Malcovati4, Max Levine1, Juan E. Arango1, Yangyu Zhou1, Francesc Solé, Catherine Cargo5, Detlef Haase6, Maria Creignou7, Ulrich Germing8, Yanming Zhang1, Gunes Gundem1, Araxe Sarian1, Arjan A. van de Loosdrecht, Martin Jädersten7, Magnus Tobiasson7, Olivier Kosmider9, Matilde Y. Follo10, Felicitas Thol11, Ronald Feitosa Pinheiro12, Valeria Santini13, Ioannis Kotsianidis14, Jacqueline Boultwood15, Fabio P.S. Santos, Julie Schanz6, Senji Kasahara, Takayuki Ishikawa, Hisashi Tsurumi16, Akifumi Takaori-Kondo2, Toru Kiguchi, Chantana Polprasert17, John M. Bennett18, Virginia M. Klimek1, Michael R. Savona19, Monika Belickova, Christina Ganster6, Laura Palomo, Guillermo Sanz20, Lionel Ades21, Matteo G. Della Porta, Alexandra Smith22, Yesenia Werner1, Minal Patel1, Agnes Viale1, Katelynd Vanness1, Donna Neuberg3, Kristen E. Stevenson3, Kamal Menghrajani1, Kelly L. Bolton1, Pierre Fenaux21, Andrea Pellagatti15, Uwe Platzbecker23, Michael Heuser11, Peter Valent24, Shigeru Chiba25, Yasushi Miyazaki26, Carlo Finelli10, Maria Teresa Voso27, Lee Yung Shih28, Michaela Fontenay9, Joop H. Jansen29, José Cervera, Yoshiko Atsuta, Norbert Gattermann8, Benjamin L. Ebert30, Rafael Bejar31, Peter L. Greenberg32, Mario Cazzola4, Eva Hellström-Lindberg7, Seishi Ogawa2, Elli Papaemmanuil1 
TL;DR: Clinical sequencing across a large prospective cohort of patients with myelodysplasic syndrome uncovers distinct associations between the mono- and biallelic states of TP53 and clinical presentation.
Abstract: Tumor protein p53 (TP53) is the most frequently mutated gene in cancer1,2. In patients with myelodysplastic syndromes (MDS), TP53 mutations are associated with high-risk disease3,4, rapid transformation to acute myeloid leukemia (AML)5, resistance to conventional therapies6–8 and dismal outcomes9. Consistent with the tumor-suppressive role of TP53, patients harbor both mono- and biallelic mutations10. However, the biological and clinical implications of TP53 allelic state have not been fully investigated in MDS or any other cancer type. We analyzed 3,324 patients with MDS for TP53 mutations and allelic imbalances and delineated two subsets of patients with distinct phenotypes and outcomes. One-third of TP53-mutated patients had monoallelic mutations whereas two-thirds had multiple hits (multi-hit) consistent with biallelic targeting. Established associations with complex karyotype, few co-occurring mutations, high-risk presentation and poor outcomes were specific to multi-hit patients only. TP53 multi-hit state predicted risk of death and leukemic transformation independently of the Revised International Prognostic Scoring System (IPSS-R)11. Surprisingly, monoallelic patients did not differ from TP53 wild-type patients in outcomes and response to therapy. This study shows that consideration of TP53 allelic state is critical for diagnostic and prognostic precision in MDS as well as in future correlative studies of treatment response. Clinical sequencing across a large prospective cohort of patients with myelodysplasic syndrome uncovers distinct associations between the mono- and biallelic states of TP53 and clinical presentation

301 citations


Journal ArticleDOI
TL;DR: Current understanding of how the bone marrow niche contributes to both the initiation and the progression of haematological malignancies is outlined and guidelines for the field are suggested which might help to overcome existing research challenges.
Abstract: Haematological malignancies were previously thought to be driven solely by genetic or epigenetic lesions within haematopoietic cells. However, the niches that maintain and regulate daily production of blood and immune cells are now increasingly being recognized as having an important role in the pathogenesis and chemoresistance of haematological malignancies. Within haematopoietic cells, the accumulation of a small number of recurrent mutations initiates malignancy. Concomitantly, specific alterations of the niches, which support haematopoietic stem cells and their progeny, can act as predisposition events, facilitating mutant haematopoietic cell survival and expansion as well as contributing to malignancy progression and providing protection of malignant cells from chemotherapy, ultimately leading to relapse. In this Perspective, we summarize our current understanding of the composition and function of the specialized haematopoietic niches of the bone marrow during health and disease. We discuss disease mechanisms (rather than malignancy subtypes) to provide a comprehensive description of key niche-associated pathways that are shared across multiple haematological malignancies. These mechanisms include primary driver mutations in bone marrow niche cells, changes associated with increased hypoxia, angiogenesis and inflammation as well as metabolic reprogramming by stromal niche cells. Consequently, remodelling of bone marrow niches can facilitate immune evasion and activation of survival pathways favouring malignant haematopoietic cell maintenance, defence against excessive reactive oxygen species and protection from chemotherapy. Lastly, we suggest guidelines for the handling and biobanking of patient samples and analysis of the niche to ensure that basic research identifying therapeutic targets can be more efficiently translated to the clinic. The hope is that integrating knowledge of how bone marrow niches contribute to haematological disease predisposition, initiation, progression and response to therapy into future clinical practice will likely improve the treatment of these disorders.

234 citations


Journal ArticleDOI
TL;DR: A vascularized human bone-marrow-on-a-chip improves the maintenance of patient-derived CD34 + cells, and recapitulates clinically relevant aspects of bone marrow injury as well as key haematopoietic defects of patients with a rare genetic disorder.
Abstract: The inaccessibility of living bone marrow (BM) hampers the study of its pathophysiology under myelotoxic stress induced by drugs, radiation or genetic mutations. Here, we show that a vascularized human BM-on-a-chip (BM chip) supports the differentiation and maturation of multiple blood cell lineages over 4 weeks while improving CD34+ cell maintenance, and that it recapitulates aspects of BM injury, including myeloerythroid toxicity after clinically relevant exposures to chemotherapeutic drugs and ionizing radiation, as well as BM recovery after drug-induced myelosuppression. The chip comprises a fluidic channel filled with a fibrin gel in which CD34+ cells and BM-derived stromal cells are co-cultured, a parallel channel lined by human vascular endothelium and perfused with culture medium, and a porous membrane separating the two channels. We also show that BM chips containing cells from patients with the rare genetic disorder Shwachman–Diamond syndrome reproduced key haematopoietic defects and led to the discovery of a neutrophil maturation abnormality. As an in vitro model of haematopoietic dysfunction, the BM chip may serve as a human-specific alternative to animal testing for the study of BM pathophysiology. A vascularized human bone-marrow-on-a-chip improves the maintenance of patient-derived CD34+ cells, and recapitulates clinically relevant aspects of bone marrow injury as well as key haematopoietic defects of patients with a rare genetic disorder.

157 citations


Journal ArticleDOI
14 May 2020-Blood
TL;DR: The spectrum of clonal hematopoiesis that can be detected in treated AML patients is discussed, terminology to standardize nomenclature in this setting is proposed, and clinical data and areas of uncertainty among the various type of post-treatment hematoietic clones are reviewed.

58 citations


Journal ArticleDOI
TL;DR: It is suggested that ≥75% LOY in bone marrow cells is associated with an increased likelihood of molecular aberrations in genes commonly seen to be altered in myeloid neoplasia and with morphological features of MDS.
Abstract: Loss of the Y chromosome (LOY) is one of the most common somatic genomic alterations in hematopoietic cells in men. However, due to the high prevalence of LOY as the sole cytogenetic finding in the healthy older population, differentiating isolated LOY associated with clonal hematologic processes from aging-associated mosaicism can be difficult in the absence of definitive morphological features of disease. In the past, various investigators have proposed that a high percentage of metaphases with LOY is more likely to represent expansion of a clonal myeloid disease-associated population. It is unknown whether the proportion of metaphases with LOY is associated with the incidence of myeloid neoplasia-associated genomic alterations. To address this question, we identified marrow samples with LOY as isolated cytogenetic finding and used targeted next generation sequencing-based molecular analysis to identify common myeloid neoplasia-associated somatic mutations. Among 73 patients with median age of 75 years (range 29-90), the percentage of metaphases with LOY was <25% in 23 patients, 25-49% in 10, 50-74% in 8 and ≥75% in 32. A threshold of ≥75% LOY was significantly associated with morphologic diagnosis of myeloid neoplasm (p = 0.004). Further, ≥75% LOY was associated with a higher lifetime incidence of diagnosis of myelodysplastic syndromes (MDS; p < 0.0001), and in multivariate analysis ≥75% LOY was a statistically significant independent predictor of myeloid neoplasia [OR 6.17; 95% CI = 2.15-17.68; p = 0.0007]. Higher LOY percentage (≥75%) was associated with greater likelihood of having somatic mutations (p = 0.0009) and a higher number of these mutations (p = 0.0002). Our findings indicate that ≥75% LOY in marrow is associated with increased likelihood of molecular alterations in genes commonly seen in myeloid neoplasia and with morphologic features of MDS. These observations suggest that ≥75% LOY in bone marrow should be considered an MDS-associated cytogenetic aberration.

31 citations


Journal ArticleDOI
17 Feb 2020-Leukemia
TL;DR: Reduced GR protein expression is implicate as a frequent cause of glucocorticoid resistance in T-ALL and CRISPR/Cas9 gene editing confirmed that loss of GR expression confers DEX resistance.
Abstract: Despite decades of clinical use, mechanisms of glucocorticoid resistance are poorly understood. We treated primary murine T lineage acute lymphoblastic leukemias (T-ALLs) with the glucocorticoid dexamethasone (DEX) alone and in combination with the pan-PI3 kinase inhibitor GDC-0941 and observed a robust response to DEX that was modestly enhanced by GDC-0941. Continuous in vivo treatment invariably resulted in outgrowth of drug-resistant clones, ~30% of which showed markedly reduced glucocorticoid receptor (GR) protein expression. A similar proportion of relapsed human T-ALLs also exhibited low GR protein levels. De novo or preexisting mutations in the gene encoding GR (Nr3c1) occurred in relapsed clones derived from multiple independent parental leukemias. CRISPR/Cas9 gene editing confirmed that loss of GR expression confers DEX resistance. Exposing drug-sensitive T-ALLs to DEX in vivo altered transcript levels of multiple genes, and this response was attenuated in relapsed T-ALLs. These data implicate reduced GR protein expression as a frequent cause of glucocorticoid resistance in T-ALL.

24 citations


Journal ArticleDOI
26 Feb 2020
TL;DR: KMT2A-MAML2 represents the first recurrent fusion described in type B thymoma and may have clinical relevance for patients with disease refractory to conventional therapeutic modalities.
Abstract: PURPOSEThymomas are epithelial neoplasms that represent the most common thymic tumors in adults. These tumors have been shown to harbor a relatively low mutational burden. As a result, there is a l...

22 citations


Journal ArticleDOI
TL;DR: It is demonstrated that although de novo and t‐ CBF‐AML patients share many features, t‐CBF‐ AML patients have worse clinical outcome than de noVO patients.
Abstract: This multi-institutional study retrospectively evaluated clinicopathologic and genetic characteristics in 351 patients with core-binding-factor acute myeloid leukemia (CBF-AML), comprising 69 therapy-related (t-CBF-AML) and 282 de novo cases. The T-CBF-AML patients were older, had lower WBC counts, and slightly higher hemoglobin than patients with de novo disease. Secondary cytogenetic abnormalities were more frequent in patients with de novo disease than t-CBF-AML (57.1% vs 41.1%, P = .026). Patients with secondary cytogenetic abnormalities had longer overall survival (OS) than those without abnormalities (median 190 vs 87 months, P = .021); trisomy 8, trisomy 22, and loss of the X or Y chromosome were associated with longer OS. In the 165 cases performed of targeted gene sequencing, pathogenic mutations were detected in 75.7% of cases, and were more frequent in de novo than in therapy-related disease (P = .013). Mutations were found in N/KRAS (37.0%), FLT3 (27.8%), KIT (17.2%), TET2 (4.9%), and ASXL1 (3.9%). The TET2 mutations were associated with shorter OS (P = .012) while N/KRAS mutation was associated with longer OS in t(8;21) AML patients (P = .001). The KIT mutation did not show prognostic significance in this cohort. Although they received similar therapy, t-CBF-AML patients had shorter OS than de novo patients (median 69 vs 190 months, P = .038). In multivariate analysis of all patients, older age and absence of any secondary cytogenetic abnormalities were significant predictors of shorter OS. Among the t-CBF-AML subset, age and hemoglobin were significant on multivariate analysis. This study demonstrated that although de novo and t-CBF-AML patients share many features, t-CBF-AML patients have worse clinical outcome than de novo patients.

22 citations


Journal ArticleDOI
TL;DR: KMT2A-fusion-positive sarcomas most commonly exhibit sclerosing epithelioid fibrosarcoma-like morphology and complex YAP1–KMT 2A–YAP1 fusions, and cases also include rare spindle-to-round cell sarcoma with VIM–K MT2A fusions and tumors of diverse histologic subtypes with unique KMT2a fusions to non-YAP 1 non-VIM partners

18 citations



Journal ArticleDOI
15 Mar 2020-Cancer
TL;DR: This phase 1 dose escalation study evaluated the safety of BV combined with mitoxantrone, etoposide, and cytarabine re‐induction chemotherapy for patients with CD30‐expressing R/R AML.
Abstract: Background Outcomes for patients with relapsed/refractory acute myeloid leukemia (R/R AML) remain poor. Novel therapies specifically targeting AML are of high interest. Brentuximab vedotin (BV) is an antibody-drug conjugate that is specific for human CD30. In this phase 1 dose escalation study, the authors evaluated the safety of BV combined with mitoxantrone, etoposide, and cytarabine (MEC) re-induction chemotherapy for patients with CD30-expressing R/R AML. Methods Using a standard dose escalation design, the authors evaluated 3 dose levels of BV (0.9 mg/kg, 1.2 mg/kg, and 1.8 mg/kg) administered once on day 1 followed by MEC on days 3 through 7. Results There were no dose-limiting toxicities noted and the maximum tolerated dose was not reached. The recommended phase 2 dose of BV was determined to be 1.8 mg/kg when combined with MEC. The side effect profile was similar to that expected from MEC chemotherapy alone, with the most common grade ≥3 toxicities being febrile neutropenia, thrombocytopenia, and anemia (toxicities were graded using the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). Among the 22 patients enrolled on the trial, the composite response rate was 36%, with a composite response rate of 42% noted among those who received the highest dose of BV. The median overall survival was 9.5 months, with a median disease-free survival of 6.8 months observed among responders. Approximately 55% of patients were able to proceed with either allogeneic hematopoietic stem cell transplantation or donor lymphocyte infusion. Conclusions The combination of BV with MEC was found to be safe in patients with CD30-expressing R/R AML and warrants further study comparing this combination with the use of MEC alone in this population (ClinicalTrials.gov identifier NCT01830777). Lay summary The outcomes for patients with relapsed/refractory acute myeloid leukemia (R/R AML) are exceptionally poor. New and emerging treatment combinations are actively being studied in an effort to improve outcomes. The authors examined the combination of brentuximab vedotin, an antibody product that recognizes a marker called CD30, with mitoxantrone, etoposide, and cytarabine (MEC), a common chemotherapy regimen, in patients with R/R AML that expressed the CD30 marker. The authors found that the combination was safe and well tolerated. Future studies comparing this new combination with the use of MEC alone can help to inform its effectiveness for this patient population.

Journal ArticleDOI
TL;DR: The hematopathologists achieved a good concordance rate of 74% in CMML vs AML classification and a high k rate, confirming that criteria for defining the blasts equivalents (blasts plus promonocytes) could be applied consistently.
Abstract: Enumeration of blasts and promonocytes is essential for World Health Organization (WHO) classification of myelomonocytic neoplasms. The accuracy of distinguishing blasts, promonocytes and monocytes, including normal vs abnormal monocytes, remains controversial. The objective of this analysis is to assess concordances between experienced hematopathologists in classifying cells as blasts, promonocytes, and monocytes according to WHO criteria. Each of 11 hematopathologists assessed glass slides from 20 patients [12 with chronic myelomonocytic leukemia (CMML) and 8 with acute myeloid leukemia (AML)] including blood and BM aspirate smears, and limited nonspecific esterase (NSE) stains. All cases were blindly reviewed. Fleiss' extension of Cohen's kappa for multiple raters was used on these variables, separately for peripheral blood (PB) and bone marrow (BM). Spearman's rank correlation was used to assess correlations between each pair of hematopathologists for each measurement. For the classification based on the sum of blasts and promonocytes in the BM, Fleiss' kappa was estimated as 0.744. For PB, categorizing patients according to the sum of blasts and promonocytes, Fleiss' kappa was estimated as 0.949. Distinction of abnormal monocytes from normal monocytes in PB did not achieve a good concordance and showed strong evidence of differences between hematopathologists (P < .0001). The hematopathologists achieved a good concordance rate of 74% in CMML vs AML classification and a high k rate, confirming that criteria for defining the blasts equivalents (blasts plus promonocytes) could be applied consistently. Identification of monocyte subtypes (abnormal vs normal) was not concordant. Our results support the practice of combining blasts/promonocytes into a single category.

Journal ArticleDOI
TL;DR: The role of pathologists in integrating data from multiple sources in the diagnosis of low-grade MDS is evolving and becoming increasingly complex; in this challenging diagnostic setting, it is important to feel comfortable with uncertainty and maintain a conservative approach.
Abstract: Objectives Myelodysplastic syndromes (MDS) are a group of myeloid neoplasms that are often difficult to diagnose due to their pathologic and clinical heterogeneity. The key features of MDS are peripheral blood cytopenias, ineffective hematopoiesis manifesting as morphologic dysplasia, and clonal genetic abnormalities. The most difficult diagnostic dilemmas often arise in low-grade MDS cases (lacking excess blasts), which can be difficult to distinguish from other causes of cytopenia. This distinction requires the integration of information from the peripheral blood (both CBC parameters and morphology), bone marrow morphology, genetic studies, and interrogation of the clinical record to exclude secondary causes. Methods We discuss the approach to the diagnosis of low-grade MDS (cases lacking increased blasts), including a diagnostic algorithm and two illustrative cases. Results The appropriate use of ancillary studies is important to support or dispute the likelihood of low-grade MDS in conjunction with the findings of morphologic dysplasia. Interpreting the results of cytogenetics and next-generation sequencing can be challenging and must incorporate the emerging knowledge of clonal hematopoiesis of indeterminate potential. Conclusions The role of pathologists in integrating data from multiple sources in the diagnosis of low-grade MDS is evolving and becoming increasingly complex; in this challenging diagnostic setting, it is important to feel comfortable with uncertainty and maintain a conservative approach.

Journal ArticleDOI
TL;DR: It is proposed that identification of the KITD816V mutation in patients diagnosed with MDS, MPN, MDS/MPN, or AML-MRC should trigger reflex testing for SM, and the SM component of their disease was not initially clinicopathologically recognized.

Journal ArticleDOI
TL;DR: Development of bone marrow (BM) fibrosis and transformation to accelerated/blast phase are the main forms of disease progression in myeloproliferative neoplasms (MPN).
Abstract: Development of bone marrow (BM) fibrosis and transformation to accelerated/blast phase are the main forms of disease progression in myeloproliferative neoplasms (MPN). Chronic myeloid leukemia, BCR-ABL1 -positive has several well-defined criteria that qualify for a diagnosis of accelerated phase (AP

Journal ArticleDOI
TL;DR: Atypical CML is a rare MDS/MPN overlap syndrome that can be diagnostically challenging; however, its emerging molecular genetic understanding and clinicomorphologic phenotype can help in distinguishing it from other Ph-negative myeloid neoplasms.
Abstract: Purpose of review Atypical chronic myeloid leukemia (aCML), BCR-ABL1-negative, is a rare myelodysplastic/myeloproliferative neoplasm (MDS/MPN) characterized by leukocytosis, granulocytic dysplasia, and typically poor patient outcomes. Since its first description as a variant CML lacking the Philadelphia chromosome (Ph), the diagnostic criteria for aCML have evolved significantly. Nevertheless, distinguishing it from other Ph-negative myeloid neoplasms can still be very challenging, and given its generally worse prognosis, this is a clinically important distinction. The purpose of this review is to conceptualize our understanding of aCML molecular genetics based on recent advances, and describe how genetic features can be used in conjunction with clinical and morphologic features to better diagnose this elusive entity. Recent findings The classification criteria for aCML have evolved and changed multiple times over the past decades, and is now based on strict application of morphologic, clinical and laboratory criteria. Recent work has elucidated the mutational landscape of aCML, especially with respect to potentially differentiating profiles compared with other Ph-negative myeloid neoplasms. Summary Atypical CML is a rare MDS/MPN overlap syndrome that can be diagnostically challenging; however, its emerging molecular genetic understanding and clinicomorphologic phenotype can help in distinguishing it from other Ph-negative myeloid neoplasms.

Journal ArticleDOI
TL;DR: Increased numbers of NPM1 wild-type cells within the microenvironment at diagnosis correlate with the subsequent presence of MRD, and Discordances between MIF and FC/NGS data suggest that aspirate materials are likely an imperfect reflection of the core biopsy tissue.

Journal ArticleDOI
TL;DR: CT-guided bone marrow procedures were more likely to result in both adequate aspirate smears and biopsy samples and longer core lengths when compared with blind procedures.
Abstract: To compare the pathology results of CT-guided and blind bone marrow aspirations and biopsies. Ninety-eight consecutive CT-guided biopsies and 98 age- and gender-matched blind (non-CT-guided) posterior iliac crest bone marrow aspirations and biopsies performed in 2017 were reviewed for adequacy of core biopsies and aspirate smears. CT procedure images and CT abdomen/pelvis images were reviewed to evaluate anatomic features of the posterior ilium and soft tissues. Statistical analysis was performed using a T test, Fisher exact test, and Kruskal-Wallis test. There was no significant difference in the age and gender of the two groups (p > 0.05). However, the CT-guided group had a higher BMI (p = 0.0049) and posterior soft tissue thickness (p = 0.0016). More CT-guided biopsy samples (CT 93 (95%); blind 77 (79%); p = 0.0006) and aspirate smears (CT 90 (92%); blind 78 (80%); p = 0.042) were categorized as adequate. The CT-guided group had longer core lengths (CT 1.4 ± 0.6 (range 0.3–3.5) cm; blind 1.0 ± 0.60 (range 0–2.6) cm; p = 0.0001). Overall, 131/164 (80%) of the cases had at least one of the described features (slanted posterior ilium (angle > 30°), 30%; rounded posterior ilium, 20%; thick posterior ilium cortex, 13%; focal lesion in posterior ilium, 12%; prior procedure in posterior ilium, 5%; posterior soft tissue thickness > 3 cm, 40%). CT-guided bone marrow procedures were more likely to result in both adequate aspirate smears and biopsy samples and longer core lengths when compared with blind procedures.



Book ChapterDOI
01 Jan 2020
TL;DR: There has been improvement in the detection of abnormal patterns and aberrant expression of antigens on all lineages in MDS with advancement of multicolor flow cytometry.
Abstract: Myelodysplastic syndromes (MDS) are a group of hematological disorders presenting with one or more clonal cytopenias, significant dyspoiesis, and increased blasts in some cases. They can be some of the most difficult diagnoses to make in both developed and developing countries. MDS is suspected by hemato-oncologists in the differential diagnosis of many cytopenic presentations and overlaps with many benign and malignant conditions. While the primary diagnosis of MDS still rests on careful morphologic assessment of blood and bone marrow, the advent of ancillary techniques such as flow cytometry, cytogenetics, and molecular genetics, including next-generation sequencing, has facilitated its diagnosis. With advancement of multicolor flow cytometry, there has been improvement in the detection of abnormal patterns and aberrant expression of antigens on all lineages in MDS. Karyotyping provides both diagnostic and prognostic information. Most recently, next generation sequencing has shed light on the molecular pathogenesis of MDS and helped in prognosticating the disease.