Network


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

Hotspot


Dive into the research topics where Haris Ali is active.

Publication


Featured researches published by Haris Ali.


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.


Biology of Blood and Marrow Transplantation | 2016

Dasatinib-Induced Colitis after Allogeneic Stem Cell Transplantation for Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia

Ibrahim Aldoss; Karl Gaal; Monzr M. Al Malki; Haris Ali; Ryotaro Nakamura; Stephen J. Forman; Vinod Pullarkat

The tyrosine kinase inhibitor dasatinib is often used after allogeneic hematopoietic cell transplantation to treat minimal residual disease in Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Colitis, sometimes hemorrhagic, has occasionally been described with the use of dasatinib for both chronic myeloid leukemia and Ph+ ALL. The pathogenesis of dasatinib-induced colitis is unclear but may be related to effects of dasatinib on immune function. We describe a series of 5 patients who had 7 episodes of colitis during dasatinib use. No patient had obvious large granular lymphocytosis in peripheral blood. The histopathologic and immunohistochemical features of these cases were indistinguishable from control cases of gut graft-versus-host disease (GVHD). In all patients symptoms resolved upon discontinuation of dasatinib in addition to therapy with local or low-dose systemic steroids. An additional 3 patients who developed cytomegalovirus (CMV) colitis while on dasatinib therapy were identified and studied. Dasatinib colitis may have an immune-mediated mechanism similar to GVHD, and dasatinib use may be associated with CMV colitis. Awareness of this association is important for avoiding unnecessary intensification of immunosuppression for suspected gut GVHD.


Nature Medicine | 2018

Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia

Bin Zhang; Le Xuan Truong Nguyen; Ling Li; Dandan Zhao; Bijender Kumar; Herman Wu; Allen Lin; Francesca Pellicano; Lisa Hopcroft; Yu-Lin Su; Mhairi Copland; Tessa L. Holyoake; Calvin J. Kuo; Ravi Bhatia; David S. Snyder; Haris Ali; Anthony S. Stein; Casey Brewer; Huafeng Wang; Tinisha McDonald; Piotr Swiderski; Estelle Troadec; Ching-Cheng Chen; Adrienne M. Dorrance; Vinod Pullarkat; Yate-Ching Yuan; Danilo Perrotti; Nadia Carlesso; Stephen J. Forman; Marcin Kortylewski

Leukemia stem cells (LSCs) in individuals with chronic myelogenous leukemia (CML) (hereafter referred to as CML LSCs) are responsible for initiating and maintaining clonal hematopoiesis. These cells persist in the bone marrow (BM) despite effective inhibition of BCR–ABL kinase activity by tyrosine kinase inhibitors (TKIs). Here we show that although the microRNA (miRNA) miR-126 supported the quiescence, self-renewal and engraftment capacity of CML LSCs, miR-126 levels were lower in CML LSCs than in long-term hematopoietic stem cells (LT-HSCs) from healthy individuals. Downregulation of miR-126 levels in CML LSCs was due to phosphorylation of Sprouty-related EVH1-domain-containing 1 (SPRED1) by BCR–ABL, which led to inhibition of the RAN–exportin-5–RCC1 complex that mediates miRNA maturation. Endothelial cells (ECs) in the BM supply miR-126 to CML LSCs to support quiescence and leukemia growth, as shown using mouse models of CML in which Mir126a (encoding miR-126) was conditionally knocked out in ECs and/or LSCs. Inhibition of BCR–ABL by TKI treatment caused an undesired increase in endogenous miR-126 levels, which enhanced LSC quiescence and persistence. Mir126a knockout in LSCs and/or ECs, or treatment with a miR-126 inhibitor that targets miR-126 expression in both LSCs and ECs, enhanced the in vivo anti-leukemic effects of TKI treatment and strongly diminished LSC leukemia-initiating capacity, providing a new strategy for the elimination of LSCs in individuals with CML.


Biology of Blood and Marrow Transplantation | 2018

Outcomes of Patients with Recurrent and Refractory Lymphoma Undergoing Allogeneic Hematopoietic Cell Transplantation with BEAM Conditioning and Sirolimus- and Tacrolimus-Based GVHD Prophylaxis

Amandeep Salhotra; Matthew Mei; Tracey Stiller; Sally Mokhtari; Alex F. Herrera; Robert Chen; Leslie Popplewell; Jasmine Zain; Haris Ali; Karamjeet Sandhu; Elizabeth Budde; Auayporn Nademanee; Stephen J. Forman; Ryotaro Nakamura

The current standard of care for patients with Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL) is high-dose conditioning followed by autologous stem cell transplantation (ASCT). For some patients (ie, those with highest-risk disease, insufficient stem cell numbers after mobilization, or bone marrow involvement) allogeneic hematopoietic cell transplantation (alloHCT) offers the potential for cure. However, the majority of patients undergoing alloHCT receive reduced-intensity conditioning as a preparative regimen, and studies assessing outcomes of patients after alloHCT with myeloablative conditioning are limited. In this retrospective study, we reviewed outcomes of 22 patients with recurrent and refractory NHL who underwent alloHCT with myeloablative BEAM conditioning and received tacrolimus/sirolimus as graft-versus-host disease (GVHD) prophylaxis at City of Hope between 2005 and 2018. With a median follow-up of 2.6 years (range, 1.0 to 11.2 years), the probabilities of 2-year overall survival and event-free survival were 58.3% (95% confidence interval [CI], 35.0% to 75.8%) and 45.5% (95% CI, 24.4% to 64.3%), respectively. The cumulative incidence of grade II to IV acute GVHD was 45.5% (95% CI, 23.8% to 64.9%), with only 1 patient developing grade IV acute GVHD. However, chronic GVHD was seen in 55% of the patients (n = 12). Of the 22 eligible patients, 2 had undergone previous ASCT and 2 had undergone previous alloHCT. Both patients with previous ASCT developed severe regimen-related toxicity. Patients who underwent alloHCT with chemorefractory disease had lower survival rates, with 1-year OS and EFS of 44.4% and 33.0%, respectively. In conclusion, alloHCT with a BEAM preparative regimen and tacrolimus/sirolimus-based GVHD should be considered as an alternative option for patients with highest-risk lymphoma whose outcomes are expectedly poor after ASCT.


Biology of Blood and Marrow Transplantation | 2017

Hyperfractionated Cyclophosphamide, Vincristine, Doxorubicin, and Dexamethasone Chemotherapy in Mantle Cell Lymphoma Patients Is Associated with Higher Rates of Hematopoietic Progenitor Cell Mobilization Failure despite Plerixafor Rescue

Amandeep Salhotra; Yuan Shan; Ni-Chun Tsai; James F. Sanchez; Ibrahim Aldoss; Haris Ali; Tanya Paris; Ricardo Spielberger; Thai M. Cao; Auayporn Nademanee; Stephen J. Forman; Robert Chen

Induction regimens for mantle cell lymphoma (MCL) can be categorized into highly intensive regimens containing cytarabine and less intense regimens, such as rituximab, cyclophosphamide, hydroxydaunorubicin, vincristine, and prednisone (R-CHOP) or rituximab with bendamustine (R-bendamustine). Prior publications have shown rituximab and hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (R-hyperCVAD) can be associated with stem cell mobilization failures. However, those studies did not include the use of plerixafor as rescue for stem cell mobilization failure. We examined our database of 181 consecutive MCL patients who received upfront therapy from 2005 to 2015 with either R-hyperCVAD or less intense chemotherapy (R-bendamustine and R-CHOP only) regimens to assess impact of frontline chemotherapy on collection of hematopoietic cell progenitors before autologous stem cell transplantation (ASCT). In the preplerixafor era (before August 16, 2009), a significant difference in peripheral blood stem cell (PBSC) collection failure between the R-hyperCVAD (12%) and other chemotherapy (11%) groups was not established. However, in the postplerixafor era, use of R-hyperCVAD chemotherapy was associated with significantly higher rates of hematopoietic progenitor cell collection failures (17%) compared with that observed in the other chemotherapy group (4%; P = .04). The rates of mobilization failure declined to 4% in the postplerixafor era from 11% in the preplerixafor era for patients receiving less intensive chemotherapy. Conversely, the rate of mobilization failure increased in the R-hyperCVAD group from 12% in the preplerixafor era to 17% in the postplerixafor era. Plerixafor does not overcome the negative impact of R-hyperCVAD on PBSC mobilization, and caution is warranted in using R-hyperCVAD in patients with newly diagnosed MCL who are candidates for ASCT.


Bone Marrow Transplantation | 2015

Mycophenolate mofetil-based salvage as acute GVHD prophylaxis after early discontinuation of tacrolimus and/or sirolimus.

Haris Ali; Joycelynne Palmer; Zeynep Eroglu; Tracey Stiller; Sandra H. Thomas; Samer K. Khaled; Sepideh Shayani; Pablo Parker; Stephen J. Forman; Ryotaro Nakamura

Mycophenolate mofetil-based salvage as acute GVHD prophylaxis after early discontinuation of tacrolimus and/or sirolimus


Blood | 2016

A Phase I Trial of Janus Kinase (JAK) Inhibition with INCB039110 in Acute Graft-Versus-Host Disease (aGVHD)

Mark A. Schroeder; H. Jean Khoury; Madan Jagasia; Haris Ali; Gary J. Schiller; Michael Arbushites; Patricia Delaite; Ying Yan; Kathleen Rhein; Miguel-Angel Perales; Yi-Bin Chen; John F. DiPersio

Collaboration


Dive into the Haris Ali'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

Anthony S. Stein

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
Top Co-Authors

Avatar

Ibrahim Aldoss

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Amandeep Salhotra

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

Monzr M. Al Malki

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Samer K. Khaled

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
Researchain Logo
Decentralizing Knowledge