Network


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

Hotspot


Dive into the research topics where Laila-Aicha Hanafi is active.

Publication


Featured researches published by Laila-Aicha Hanafi.


Journal of Clinical Oncology | 2017

Durable Molecular Remissions in Chronic Lymphocytic Leukemia Treated With CD19-Specific Chimeric Antigen Receptor–Modified T Cells After Failure of Ibrutinib

Cameron J. Turtle; Kevin A. Hay; Laila-Aicha Hanafi; Daniel Li; Sindhu Cherian; Xueyan Chen; Brent L. Wood; Arletta Lozanski; John C. Byrd; Shelly Heimfeld; Stanley R. Riddell; David G. Maloney

Purpose We evaluated the safety and feasibility of anti-CD19 chimeric antigen receptor-modified T (CAR-T) cell therapy in patients with chronic lymphocytic leukemia (CLL) who had previously received ibrutinib. Methods Twenty-four patients with CLL received lymphodepleting chemotherapy and anti-CD19 CAR-T cells at one of three dose levels (2 × 105, 2 × 106, or 2 × 107 CAR-T cells/kg). Nineteen patients experienced disease progression while receiving ibrutinib, three were ibrutinib intolerant, and two did not experience progression while receiving ibrutinib. Six patients were venetoclax refractory, and 23 had a complex karyotype and/or 17p deletion. Results Four weeks after CAR-T cell infusion, the overall response rate (complete response [CR] and/or partial response [PR]) by International Workshop on Chronic Lymphocytic Leukemia (IWCLL) criteria was 71% (17 of 24). Twenty patients (83%) developed cytokine release syndrome, and eight (33%) developed neurotoxicity, which was reversible in all but one patient with a fatal outcome. Twenty of 24 patients received cyclophosphamide and fludarabine lymphodepletion and CD19 CAR-T cells at or below the maximum tolerated dose (≤ 2 × 106 CAR-T cells/kg). In 19 of these patients who were restaged, the overall response rate by IWCLL imaging criteria 4 weeks after infusion was 74% (CR, 4/19, 21%; PR, 10/19, 53%), and 15/17 patients (88%) with marrow disease before CAR-T cells had no disease by flow cytometry after CAR-T cells. Twelve of these patients underwent deep IGH sequencing, and seven (58%) had no malignant IGH sequences detected in marrow. Absence of the malignant IGH clone in marrow of patients with CLL who responded by IWCLL criteria was associated with 100% progression-free survival and overall survival (median 6.6 months follow-up) after CAR-T cell immunotherapy. The progression-free survival was similar in patients with lymph node PR or CR by IWCLL criteria. Conclusion CD19 CAR-T cells are highly effective in high-risk patients with CLL after they experience treatment failure with ibrutinib therapy.


Cancer Discovery | 2017

Endothelial Activation and Blood-Brain Barrier Disruption in Neurotoxicity after Adoptive Immunotherapy with CD19 CAR-T Cells.

Juliane Gust; Kevin A. Hay; Laila-Aicha Hanafi; Daniel Li; David Myerson; Luis F. Gonzalez-Cuyar; Cecilia Yeung; W. Conrad Liles; Mark M. Wurfel; José A. López; Junmei Chen; Dominic W. Chung; Susanna Harju-Baker; Tahsin Özpolat; Kathleen R. Fink; Stanley R. Riddell; David G. Maloney; Cameron J. Turtle

Lymphodepletion chemotherapy followed by infusion of CD19-targeted chimeric antigen receptor-modified T (CAR-T) cells can be complicated by neurologic adverse events (AE) in patients with refractory B-cell malignancies. In 133 adults treated with CD19 CAR-T cells, we found that acute lymphoblastic leukemia, high CD19+ cells in bone marrow, high CAR-T cell dose, cytokine release syndrome, and preexisting neurologic comorbidities were associated with increased risk of neurologic AEs. Patients with severe neurotoxicity demonstrated evidence of endothelial activation, including disseminated intravascular coagulation, capillary leak, and increased blood-brain barrier (BBB) permeability. The permeable BBB failed to protect the cerebrospinal fluid from high concentrations of systemic cytokines, including IFNγ, which induced brain vascular pericyte stress and their secretion of endothelium-activating cytokines. Endothelial activation and multifocal vascular disruption were found in the brain of a patient with fatal neurotoxicity. Biomarkers of endothelial activation were higher before treatment in patients who subsequently developed grade ≥4 neurotoxicity.Significance: We provide a detailed clinical, radiologic, and pathologic characterization of neurotoxicity after CD19 CAR-T cells, and identify risk factors for neurotoxicity. We show endothelial dysfunction and increased BBB permeability in neurotoxicity and find that patients with evidence of endothelial activation before lymphodepletion may be at increased risk of neurotoxicity. Cancer Discov; 7(12); 1404-19. ©2017 AACR.See related commentary by Mackall and Miklos, p. 1371This article is highlighted in the In This Issue feature, p. 1355.


Blood | 2017

Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor–modified T-cell therapy

Kevin A. Hay; Laila-Aicha Hanafi; Daniel Li; Juliane Gust; Wc Liles; Mark M. Wurfel; José A. López; Junmei Chen; Dominic W. Chung; Susanna Harju-Baker; Sindhu Cherian; Xiaoji Chen; Riddell; David G. Maloney; Cameron J. Turtle

Lymphodepletion chemotherapy followed by infusion of CD19-specific chimeric antigen receptor-modified (CAR) T cells has produced impressive antitumor responses in patients with refractory CD19+ B-cell malignancies but is often associated with cytokine release syndrome (CRS). Our understanding of CRS continues to evolve, and identification of the kinetics of CRS and predictive clinical and laboratory biomarkers of severity are needed to evaluate strategies to mitigate toxicity. We report the clinical presentation of and identify biomarkers of severe CRS in 133 adult patients who received CD19 CAR T cells. CRS developed in 70% of patients, including 62.5% with grade 1 to 3 CRS (grade 1, 26%; grade 2, 32%; grade 3, 4.5%), 3.8% with grade 4, and 3.8% with grade 5. A majority of cases of grade ≥4 CRS occurred during CAR T-cell dose finding. Multivariable analysis of baseline characteristics identified high marrow tumor burden, lymphodepletion using cyclophosphamide and fludarabine, higher CAR T-cell dose, thrombocytopenia before lymphodepletion, and manufacturing of CAR T cells without selection of CD8+ central memory T cells as independent predictors of CRS. Severe CRS was characterized by hemodynamic instability, capillary leak, and consumptive coagulopathy. Angiopoietin-2 and von Willebrand factor, which are biomarkers of endothelial activation, were increased during severe CRS and also before lymphodepletion in patients who subsequently developed CRS. We describe a classification-tree algorithm to guide studies of early intervention after CAR T-cell infusion for patients at high risk of severe CRS. These data provide a framework for early intervention studies to facilitate safer application of effective CD19 CAR T-cell therapy.


Biology of Blood and Marrow Transplantation | 2018

Graft-Derived Reconstitution of Mucosal-Associated Invariant T Cells after Allogeneic Hematopoietic Cell Transplantation

Abir Bhattacharyya; Laila-Aicha Hanafi; Alyssa Sheih; Jonathan L. Golob; Sujatha Srinivasan; Michael Boeckh; Steven A. Pergam; Sajid Mahmood; Kelsey K. Baker; Ted Gooley; Filippo Milano; David N. Fredricks; Stanley R. Riddell; Cameron J. Turtle

Mucosal-associated invariant T (MAIT) cells express a semi-invariant Vα7.2+ T cell receptor (TCR) that recognizes ligands from distinct bacterial and fungal species. In neonates, MAIT cells proliferate coincident with gastrointestinal (GI) bacterial colonization. In contrast, under noninflammatory conditions adult MAIT cells remain quiescent because of acquired regulation of TCR signaling. Effects of inflammation and the altered GI microbiota after allogeneic hematopoietic cell transplantation (HCT) on MAIT cell reconstitution have not been described. We conducted an observational study of MAIT cell reconstitution in myeloablative (n = 41) and nonmyeloablative (n = 66) allogeneic HCT recipients and found that despite a rapid and early increase to a plateau at day 30 after HCT, MAIT cell numbers failed to normalize for at least 1 year. Cord blood transplant recipients and those who received post-HCT cyclophosphamide for graft versus host disease (GVHD) prophylaxis had profoundly impaired MAIT cell reconstitution. Sharing of TCRβ gene sequences between MAIT cells isolated from HCT grafts and blood of recipients after HCT showed early MAIT cell reconstitution was due at least in part to proliferation of MAIT cells transferred in the HCT graft. Inflammatory cytokines were required for TCR-dependent MAIT cell proliferation, suggesting that bacterial Vα7.2+ TCR ligands might promote MAIT cell reconstitution after HCT. Robust MAIT cell reconstitution was associated with an increased GI abundance of Blautia spp. MAIT cells suppressed proliferation of conventional T cells consistent with a possible regulatory role. Our data identify modifiable factors impacting MAIT cell reconstitution that could influence the risk of GVHD after HCT.


Nature Biotechnology | 2017

HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells

Germán G Gornalusse; Roli K. Hirata; Sarah E. Funk; Laura Riolobos; Vanda S Lopes; Gabriel Manske; Donna Prunkard; Aric G Colunga; Laila-Aicha Hanafi; Dennis O. Clegg; Cameron J. Turtle; David W. Russell


Blood | 2015

Anti-CD19 Chimeric Antigen Receptor-Modified T Cell Therapy for B Cell Non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia: Fludarabine and Cyclophosphamide Lymphodepletion Improves In Vivo Expansion and Persistence of CAR-T Cells and Clinical Outcomes

Cameron J. Turtle; Carolina Berger; Daniel Sommermeyer; Laila-Aicha Hanafi; Barbara Pender; Emily Robinson; Katherine Melville; Tanya M Budiarto; Natalia N Steevens; Colette Chaney; Sindhu Cherian; Brent L. Wood; Lorinda Soma; Xueyan Chen; Shelly Heimfeld; Michael C. Jensen; Stanley R. Riddell; David G. Maloney


Blood | 2015

Addition of Fludarabine to Cyclophosphamide Lymphodepletion Improves In Vivo Expansion of CD19 Chimeric Antigen Receptor-Modified T Cells and Clinical Outcome in Adults with B Cell Acute Lymphoblastic Leukemia

Cameron J. Turtle; Laila-Aicha Hanafi; Carolina Berger; Daniel Sommermeyer; Barbara Pender; Emily Robinson; Katherine Melville; Tanya M Budiarto; Natalia N Steevens; Colette Chaney; Sindhu Cherian; Brent L. Wood; Lorinda Soma; Xueyan Chen; Shelly Heimfeld; Michael C. Jensen; Stanley R. Riddell; David G. Maloney


Journal of Clinical Oncology | 2016

Rate of durable complete response in ALL, NHL, and CLL after immunotherapy with optimized lymphodepletion and defined composition CD19 CAR-T cells.

Cameron J. Turtle; Laila-Aicha Hanafi; Carolina Berger; Theodore A. Gooley; Colette Chaney; Sindhu Cherian; Lori Soma; Xueyan Chen; Cecilia Cs Yeung; Keith R. Loeb; Brent L. Wood; Michael Hudecek; Daniel Sommermeyer; Daniel Li; Kevin A. Hay; Shelly Heimfeld; Stanley R. Riddell; David G. Maloney


Journal of Clinical Oncology | 2015

Immunotherapy with CD19-specific chimeric antigen receptor (CAR)-modified T cells of defined subset composition.

Cameron J. Turtle; Carolina Berger; Daniel Sommermeyer; Tanya M Budiarto; Laila-Aicha Hanafi; Katherine Melville; Barbara Pender; Natalia N Steevens; Colette Chaney; Shelly Heimfeld; Sindhu Cherian; Brent L. Wood; Lori Soma; Xueyan Chen; Michael C. Jensen; Stan R. Riddell; David G. Maloney


Journal of Clinical Oncology | 2017

Cytokine release syndrome (CRS) and neurotoxicity (NT) after CD19-specific chimeric antigen receptor- (CAR-) modified T cells.

Cameron J. Turtle; Kevin Anthony Hay; Juliane Gust; Laila-Aicha Hanafi; Daniel Li; W. Conrad Liles; Mark M. Wurfel; Susanna Harju-Baker; David Myerson; Luis F. Gonzalez-Cuyar; Cecilia Cs Yeung; Stanley R. Riddell; David G. Maloney

Collaboration


Dive into the Laila-Aicha Hanafi's collaboration.

Top Co-Authors

Avatar

Cameron J. Turtle

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

David G. Maloney

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Colette Chaney

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Sindhu Cherian

University of Washington Medical Center

View shared research outputs
Top Co-Authors

Avatar

Brent L. Wood

University of Washington Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xueyan Chen

University of Washington Medical Center

View shared research outputs
Top Co-Authors

Avatar

Carolina Berger

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Kevin A. Hay

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge