Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Samer K. Khaled is active.

Publication


Featured researches published by Samer K. Khaled.


Blood | 2016

Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL.

Xiuli Wang; Leslie Popplewell; Jamie R. Wagner; Araceli Naranjo; M. Suzette Blanchard; Michelle Mott; Adam P. Norris; Ching Lam W Wong; Ryan Urak; Wen-Chung Chang; Samer K. Khaled; Tanya Siddiqi; Lihua E. Budde; Jingying Xu; Brenda Chang; Nikita Gidwaney; Sandra H. Thomas; Laurence J.N. Cooper; Stanley R. Riddell; Christine E. Brown; Michael C. Jensen; Stephen J. Forman

Myeloablative autologous hematopoietic stem cell transplantation (HSCT) is a mainstay of therapy for relapsed intermediate-grade B-cell non-Hodgkin lymphoma (NHL); however, relapse rates are high. In phase 1 studies designed to improve long-term remission rates, we administered adoptive T-cell immunotherapy after HSCT, using ex vivo-expanded autologous central memory-enriched T cells (TCM) transduced with lentivirus expressing CD19-specific chimeric antigen receptors (CARs). We present results from 2 safety/feasibility studies, NHL1 and NHL2, investigating different T-cell populations and CAR constructs. Engineered TCM-derived CD19 CAR T cells were infused 2 days after HSCT at doses of 25 to 200 × 10(6) in a single infusion. In NHL1, 8 patients safely received T-cell products engineered from enriched CD8(+) TCM subsets, expressing a first-generation CD19 CAR containing only the CD3ζ endodomain (CD19R:ζ). Four of 8 patients (50%; 95% confidence interval [CI]: 16-84%) were progression free at both 1 and 2 years. In NHL2, 8 patients safely received T-cell products engineered from enriched CD4(+) and CD8(+) TCM subsets and expressing a second-generation CD19 CAR containing the CD28 and CD3ζ endodomains (CD19R:28ζ). Six of 8 patients (75%; 95% CI: 35-97%) were progression free at 1 year. The CD4(+)/CD8(+) TCM-derived CD19 CAR T cells (NHL2) exhibited improvement in expansion; however, persistence was ≤28 days, similar to that seen by others using CD28 CARs. Neither cytokine release syndrome nor delayed hematopoietic engraftment was observed in either trial. These data demonstrate the safety and feasibility of CD19 CAR TCM therapy after HSCT. Trials were registered at www.clinicaltrials.gov as #NCT01318317 and #NCT01815749.


Current Opinion in Oncology | 2012

Allogeneic hematopoietic cell transplantation for acute lymphoblastic leukemia in adults

Samer K. Khaled; Sandra H. Thomas; Stephen J. Forman

Purpose of review Acute lymphoblastic leukemia (ALL) is a heterogeneous disease, for which treatment guidelines are still evolving. Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative therapeutic modality for ALL, and this review describes the recent studies and current practice patterns concerning the who, when, and how of allo-HCT in the management of ALL. Recent findings Allogeneic stem cell transplantation is the treatment of choice for patients with ALL after first relapse and is also recommended for high-risk patients in first complete remission (CR1). Minimal residual disease evaluation and monitoring is developing as an important prognostic factor and could guide physicians in determining which patients, especially those with standard risk, might require transplant. Tyrosine kinase inhibitor (TKI) therapy allows a much higher proportion of Philadelphia-chromosome-positive ALL patients to attain remission and proceed to transplant with improved results; posttransplant TKI maintenance therapy may also provide survival benefit. Reduced-intensity conditioning regimens are a reasonable alternative for patients who would otherwise be ineligible for transplant because of age or comorbidity. Summary For patients with high-risk features, there is general agreement that allo-HCT in CR1 is a potentially curative option; however, there is no consensus on early transplant for standard-risk patients.


British Journal of Haematology | 2013

Criteria for and outcomes of allogeneic haematopoietic stem cell transplant in children, adolescents and young adults with acute lymphoblastic leukaemia in first complete remission

Jessica Hochberg; Samer K. Khaled; Stephen J. Forman; Mitchell S. Cairo

Most children, adolescents and young adults with acute lymphoblastic leukaemia (ALL) in first complete remission (CR1) have an excellent prognosis with multi‐agent chemotherapy in induction, consolidation, re‐induction and maintenance therapy. However, there is a subset of patients with a more guarded prognosis using this approach, who may benefit from haematopoietic allogeneic stem cell transplantation (alloHSCT). Commonly used criteria for alloHSCT in children, adolescents and young adults with ALL in CR1 include: induction failure, poor cytogenetics, persistent minimal residual disease (MRD), age, immunophenotype, white blood cell count at diagnosis and rapidity of induction response. Two‐year event‐free survival following alloHSCT in patients with ALL in CR1 ranges from 50 to 80% depending on disease status, donor source, conditioning therapy, age and other risk factors. Future studies should focus on more precisely identifying poor‐risk features, such as disease genomics and host pharmacogenomics, refining MRD measurements, improving unrelated donor matching, reducing MRD prior to alloHSCT, and developing post‐alloHSCT humoral and cellular therapy approaches.


Bone Marrow Transplantation | 2013

A phase II study of sirolimus, tacrolimus and rabbit anti-thymocyte globulin as GVHD prophylaxis after unrelated-donor PBSC transplant.

Samer K. Khaled; Joycelynne Palmer; Tracey Stiller; David Senitzer; Maegawa R; Roberto Rodriguez; Pablo Parker; Auayporn Nademanee; Cai Jl; David S. Snyder; Chatchada Karanes; Osorio E; Sandra H. Thomas; Stephen J. Forman; Ryotaro Nakamura

We report on a prospective phase II trial of 32 patients who underwent unrelated-donor hematopoietic cell transplantation, with a tacrolimus, sirolimus and rabbit anti-thymoctye globulin GVHD prophylactic regimen. The primary study endpoint was incidence of grades II–IV acute (aGVHD), with 80% power to detect a 30% decrease compared with institutional historical controls. Median age at transplant was 60 (19–71). In total, 23 patients (72%) received reduced-intensity conditioning, whereas the remainder received full-intensity regimens. Median follow-up for surviving patients was 35 months (range: 21–49). The cumulative incidence of aGVHD was 37.3%, and the 2-year cumulative incidence of chronic GVHD was 63%. We observed thrombotic microangiopathy in seven patients (21.8%), one of whom also developed sinusoidal obstructive syndrome (SOS). Four of the 32 patients (12.5%) failed to engraft, and 3 of these 4 died. As a result, enrollment to this trial was closed before the targeted accrual of 60 patients. Two-year OS was 65.5% and EFS was 61.3%. Two-year cumulative incidence of relapse was 12.5% and non-relapse mortality (NRM) was 15.6%. NRM and aGVHD rates were lower than historical rates. However, the unexpectedly high incidence of graft failure requires caution in the design of future studies with this regimen.


American Journal of Hematology | 2018

Results of a phase 1 study of quizartinib as maintenance therapy in subjects with acute myeloid leukemia in remission following allogeneic hematopoietic stem cell transplant.

Samer K. Khaled; Betul Oran; Guy Gammon; Denise Trone; Olga Frankfurt

FLT3‐ITD–mutated acute myeloid leukemia (AML) has very high risk of relapse and is associated with poor outcome following allogeneic hematopoietic‐cell transplant (allo‐HCT). This two‐part, phase 1, multicenter, open‐label, sequential‐group, dose‐escalation study aimed to determine dose‐limiting toxicities (DLTs), maximum tolerated dose (MTD), and safety/tolerability of quizartinib, a selective and highly potent FLT3 inhibitor, when administered as maintenance therapy after allo‐HCT. Thirteen subjects with documented FLT3‐ITD–mutated AML in morphological remission following allo‐HCT received one of two quizartinib dihydrochloride dose levels (DL): 40 mg/d (DL1; n = 7) and 60 mg/d (DL2; n = 6), administered orally in 28‐day cycles for up to 24 cycles. Median age of participants was 43 years. All subjects received human leukocyte antigen (HLA)‐matched allo‐HCT. One subject treated at DL1 and 1 treated at DL2 had DLTs that required drug interruption (grade 3 gastric hemorrhage and grade 3 anemia, respectively). Ten subjects (77%) received quizartinib for >1 year; 5 (38%) completed 24 cycles. Four subjects (31%) discontinued quizartinib due to adverse events. One subject (8%) experienced relapse during cycle 1 and discontinued treatment. Most common grade 3/4 adverse events were neutropenia (23%), anemia (15%), leukopenia (15%), lymphopenia (15%), and thrombocytopenia (15%). This study demonstrated acceptable tolerability and early evidence of reduced relapse rate following allo‐HCT with quizartinib maintenance compared to historical cohorts. No MTD was identified, but 60 mg daily was selected as highest dose for continuous daily administration based on randomized comparison of daily 30 and 60 mg doses in relapsed/refractory AML.


Biology of Blood and Marrow Transplantation | 2017

Phase I Trial of Total Marrow and Lymphoid Irradiation Transplantation Conditioning in Patients with Relapsed/Refractory Acute Leukemia

Anthony S. Stein; Joycelynne Palmer; Ni-Chun Tsai; Monzr M. Al Malki; Ibrahim Aldoss; Haris Ali; Ahmed Aribi; Len Farol; Chatchada Karanes; Samer K. Khaled; An Liu; Margaret R. O'Donnell; Pablo Parker; Anna Pawlowska; Vinod Pullarkat; Eric H. Radany; Joseph Rosenthal; Firoozeh Sahebi; Amandeep Salhotra; James F. Sanchez; Tim Schultheiss; Ricardo Spielberger; Sandra H. Thomas; David S. Snyder; Ryotaro Nakamura; Guido Marcucci; Stephen J. Forman; Jeffrey Y.C. Wong

Current conditioning regimens provide insufficient disease control in relapsed/refractory acute leukemia patients undergoing hematopoietic stem cell transplantation (HSCT) with active disease. Intensification of chemotherapy and/or total body irradiation (TBI) is not feasible because of excessive toxicity. Total marrow and lymphoid irradiation (TMLI) allows for precise delivery and increased intensity treatment via sculpting radiation to sites with high disease burden or high risk for disease involvement, while sparing normal tissue. We conducted a phase I trial in 51 patients (age range, 16 to 57 years) with relapsed/refractory acute leukemia undergoing HSCT (matched related, matched unrelated, or 1-allele mismatched unrelated) with active disease, combining escalating doses of TMLI (range, 1200 to 2000 cGy) with cyclophosphamide (CY) and etoposide (VP16). The maximum tolerated dose was declared at 2000 cGy, as TMLI simulation studies indicated that >2000 cGy might deliver doses toxic for normal organs at or exceeding those delivered by standard TBI. The post-transplantation nonrelapse mortality (NRM) rate was only 3.9% (95% confidence interval [CI], .7 to 12.0) at day +100 and 8.1% (95% CI, 2.5 to 18.0) at 1 year. The cumulative incidence of grades II to IV acute graft-versus-host disease (GVHD) was 43.1% (95% CI, 29.2 to 56.3) and for grade III and IV, it was 13.7% (95% CI, 6.9 to 27.3). The day +30 complete remission rate for all patients was 88% and was 100% for those treated at 2000 cGy. The overall 1-year survival was 55.5% (95% CI, 40.7 to 68.1). The TMLI/CY/VP16 conditioning regimen is well tolerated at TMLI doses up to 2000 cGy with a low 100-day and 1-year NRM rate and no increased risk of GVHD with higher doses of radiation.


Haematologica | 2017

Favorable impact of allogeneic stem cell transplantation in patients with therapy-related myelodysplasia regardless of TP53 mutational status

Ibrahim Aldoss; Anh Pham; Sierra Min Li; Ketevan Gendzekhadze; Michelle Afkhami; Mihan Telatar; Hao Hong; Abbas Padeganeh; Victoria Bedell; Thai Cao; Samer K. Khaled; Monzr M. Al Malki; Amandeep Salhotra; Haris Ali; Ahmed Aribi; Joycelynne Palmer; Patricia Aoun; Ricardo Spielberger; Anthony S. Stein; David S. Snyder; Margaret R. O'Donnell; Joyce Murata-Collins; David Senitzer; Dennis D. Weisenburger; Stephen J. Forman; Vinod Pullarkat; Guido Marcucci; Raju Pillai; Ryotaro Nakamura

Therapy-related myelodysplastic syndrome is a long-term complication of cancer treatment in patients receiving cytotoxic therapy, characterized by high-risk genetics and poor outcomes. Allogeneic hematopoietic cell transplantation is the only potential cure for this disease, but the prognostic impact of pre-transplant genetics and clinical features has not yet been fully characterized. We report here the genetic and clinical characteristics and outcomes of a relatively large cohort of patients with therapy-related myelodysplastic syndrome (n=67) who underwent allogeneic transplantation, comparing these patients to similarly treated patients with de novo disease (n=199). The 5-year overall survival was not different between patients with therapy-related and de novo disease (49.9% versus 53.9%; P=0.61) despite a higher proportion of individuals with an Intermediate-2/High International Prognostic Scoring System classification (59.7% versus 43.7%; P=0.003) and high-risk karyotypes (61.2% versus 30.7%; P<0.01) among the patients with therapy-related disease. In mutational analysis, TP53 alteration was the most common abnormality in patients with therapy-related disease (n=18: 30%). Interestingly, the presence of mutations in TP53 or in any other of the high-risk genes (EZH2, ETV6, RUNX1, ASXL1: n=29: 48%) did not significantly affect either overall survival or relapse-free survival. Allogeneic stem-cell transplantation is, therefore, a curative treatment for patients with therapy-related myelodysplastic syndrome, conferring a similar long-term survival to that of patients with de novo disease despite higher-risk features. While TP53 alteration was the most common mutation in therapy-related myelodysplastic syndrome, the finding was not detrimental in our case-series.


Haematologica | 2018

Therapy-related acute lymphoblastic leukemia has distinct clinical and cytogenetic features compared to de novo acute lymphoblastic leukemia, but outcomes are comparable in transplanted patients

Ibrahim Aldoss; Tracey Stiller; Ni-Chun Tsai; Joo Y. Song; Thai Cao; N. Achini Bandara; Amadeep Salhotra; Samer K. Khaled; Ahmed Aribi; Monzr M. Al Malki; Matthew Mei; Haris Ali; Ricardo Spielberger; Margaret R. O'Donnell; David S. Snyder; Thomas P. Slavin; Ryotaro Nakamura; Anthony S. Stein; Stephen J. Forman; Guido Marcucci; Vinod Pullarkat

Therapy-related acute lymphoblastic leukemia remains poorly defined due to a lack of large data sets recognizing the defining characteristics of this entity. We reviewed all consecutive cases of adult acute lymphoblastic leukemia treated at our institution between 2000 and 2017 and identified therapy-related cases - defined as acute lymphoblastic leukemia preceded by prior exposure to cytotoxic chemotherapy and/or radiation. Of 1022 patients with acute lymphoblastic leukemia, 93 (9.1%) were classified as therapy-related. The median latency for therapy-related acute lymphoblastic leukemia onset was 6.8 years from original diagnosis, and this was shorter for patients carrying the MLL gene rearrangement compared to those with other cytogenetics. When compared to de novo acute lymphoblastic leukemia, therapy-related patients were older (P<0.01), more often female (P<0.01), and had more MLL gene rearrangement (P<0.0001) and chromosomes 5/7 aberrations (P=0.02). Although therapy-related acute lymphoblastic leukemia was associated with inferior 2-year overall survival compared to de novo cases (46.0% vs. 68.1%, P=0.001), prior exposure to cytotoxic therapy (therapy-related) did not independently impact survival in multivariate analysis (HR=1.32; 95% CI: 0.97–1.80, P=0.08). There was no survival difference (2-year = 53.4% vs. 58.9%, P=0.68) between the two groups in patients who received allogenic hematopoietic cell transplantation. In conclusion, therapy-related acute lymphoblastic leukemia represents a significant proportion of adult acute lymphoblastic leukemia diagnoses, and a subset of cases carry clinical and cytogenetic abnormalities similar to therapy-related myeloid neoplasms. Although survival of therapy-related acute lymphoblastic leukemia was inferior to de novo cases, allogeneic hematopoietic cell transplantation outcomes were comparable for the two entities.


American Journal of Hematology | 2017

Philadelphia chromosome as a recurrent event among therapy-related acute leukemia: ALDOSS et al.

Ibrahim Aldoss; Tracey Stiller; Joo Y. Song; Monzr M. Al Malki; Haris Ali; Amandeep Salhotra; Ahmed Aribi; Samer K. Khaled; Popsie Gaytan; Joyce Murata-Collins; Joycelynne Palmer; David S. Snyder; Margaret R. O'Donnell; Ryotaro Nakamura; Anthony S. Stein; Stephen J. Forman; Guido Marcucci; Vinod Pullarkat

long-term outcome of NMP1 mutation positive patients transplanted after relapse. In conclusion, our single-institution experience supports the reported superior shortterm prognosis for patients with NPM1 mutated AML, but challenges the accepted notion of favorable long-term outcome in this population. Prospective clinical trials are needed to clarify long-term prognosis and optimal management of these patients.


Cell Stem Cell | 2018

SIRT1 Activation Disrupts Maintenance of Myelodysplastic Syndrome Stem and Progenitor Cells by Restoring TET2 Function

Jie Sun; Xin He; Yinghui Zhu; Zonghui Ding; Haojie Dong; Yimei Feng; Juan Du; Hanying Wang; Xiwei Wu; Lei Zhang; Xiaochun Yu; Allen Lin; Tinisha McDonald; Dandan Zhao; Herman Wu; Wei-Kai Hua; Bin Zhang; Lifeng Feng; Kaoru Tohyama; Ravi Bhatia; Philipp Oberdoerffer; Yang Jo Chung; Peter D. Aplan; Jacqueline Boultwood; Andrea Pellagatti; Samer K. Khaled; Marcin Kortylewski; Flavia Pichiorri; Ya-Huei Kuo; Nadia Carlesso

Myelodysplastic syndrome (MDS), a largely incurable hematological malignancy, is derived from aberrant clonal hematopoietic stem/progenitor cells (HSPCs) that persist after conventional therapies. Defining the mechanisms underlying MDS HSPC maintenance is critical for developing MDS therapy. The deacetylase SIRT1 regulates stem cell proliferation, survival, and self-renewal by deacetylating downstream proteins. Here we show that SIRT1 protein levels were downregulated in MDS HSPCs. Genetic or pharmacological activation of SIRT1 inhibited MDS HSPC functions, whereas SIRT1 deficiency enhanced MDS HSPC self-renewal. Mechanistically, the inhibitory effects of SIRT1 were dependent on TET2, a safeguard against HSPC transformation. SIRT1 deacetylated TET2 at conserved lysine residues in its catalytic domain, enhancing TET2 activity. Our genome-wide analysis identified cancer-related genes regulated by the SIRT1/TET2 axis. SIRT1 activation also inhibited functions of MDS HSPCs from patients with TET2 heterozygous mutations. Altogether, our results indicate that restoring TET2 function through SIRT1 activation represents a promising means to target MDS HSPCs.

Collaboration


Dive into the Samer K. Khaled's collaboration.

Top Co-Authors

Avatar

Stephen J. Forman

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ryotaro Nakamura

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joycelynne Palmer

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

David S. Snyder

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Vinod Pullarkat

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anthony S. Stein

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Haris Ali

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sandra H. Thomas

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Senitzer

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Guido Marcucci

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge