Network


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

Hotspot


Dive into the research topics where Antonio Cavazos is active.

Publication


Featured researches published by Antonio Cavazos.


Clinical Cancer Research | 2017

Antileukemia efficacy and mechanisms of action of SL-101, a novel anti-CD123 antibody conjugate, in acute myeloid leukemia

Lina Han; Jeffrey L. Jorgensen; Christopher Brooks; Ce Shi; Qi Zhang; Graciela Nogueras Gonzalez; Antonio Cavazos; Rongqing Pan; Hong Mu; Sa A. Wang; Jin Zhou; Gheath AI-Atrash; Stefan O. Ciurea; Michael P. Rettig; John F. DiPersio; Jorge Cortes; Xuelin Huang; Hagop M. Kantarjian; Michael Andreeff; Farhad Ravandi; Marina Konopleva

Purpose: The persistence of leukemia stem cells (LSC)-containing cells after induction therapy may contribute to minimal residual disease (MRD) and relapse in acute myeloid leukemia (AML). We investigated the clinical relevance of CD34+CD123+ LSC-containing cells and antileukemia potency of a novel antibody conjugate SL-101 in targeting CD123+ LSCs. Experimental Methods and Results: In a retrospective study on 86 newly diagnosed AML patients, we demonstrated that a higher proportion of CD34+CD123+ LSC-containing cells in remission was associated with persistent MRD and predicted shorter relapse-free survival in patients with poor-risk cytogenetics. Using flow cytometry, we explored the potential benefit of therapeutic targeting of CD34+CD38−CD123+ cells by SL-101, a novel antibody conjugate comprising an anti-CD123 single-chain Fv fused to Pseudomonas exotoxin A. The antileukemia potency of SL-101 was determined by the expression levels of CD123 antigen in a panel of AML cell lines. Colony-forming assay established that SL-101 strongly and selectively suppressed the function of leukemic progenitors while sparing normal counterparts. The internalization, protein synthesis inhibition, and flow cytometry assays revealed the mechanisms underlying the cytotoxic activities of SL-101 involved rapid and efficient internalization of antibody, sustained inhibition of protein synthesis, induction of apoptosis, and blockade of IL3-induced p-STAT5 and p-AKT signaling pathways. In a patient-derived xenograft model using NSG mice, the repopulating capacity of LSCs pretreated with SL-101 in vitro was significantly impaired. Conclusions: Our data define the mechanisms by which SL-101 targets AML and warrant further investigation of the clinical application of SL-101 and other CD123-targeting strategies in AML. Clin Cancer Res; 23(13); 3385–95. ©2017 AACR.


Oncotarget | 2018

Inhibition of mTORC1/C2 signaling improves anti-leukemia efficacy of JAK/STAT blockade in CRLF2 rearranged and/or JAK driven philadelphia chromosome-like acute B-cell lymphoblastic leukemia

Qi Zhang; Ce Shi; Lina Han; Nitin Jain; Kathryn G. Roberts; Helen Ma; Tianyu Cai; Antonio Cavazos; Yoko Tabe; Rodrigo Jacamo; Hong Mu; Yang Zhao; Jing Wang; Shuo Chieh Wu; Fenglin Cao; Zhihong Zeng; Jin Zhou; Yingchang Mi; Elias Jabbour; Ross L. Levine; Sarah K. Tasian; Charles G. Mullighan; David M. Weinstock; David A. Fruman; Marina Konopleva

Patients with cytokine receptor-like factor 2 rearranged (CRLF2-re) subgroup Philadelphia chromosome–like B-cell acute lymphoblastic leukemia (Ph-like B-ALL) have a high relapse rate and poor clinical outcomes. CRFL2-re Ph-like B-ALL is characterized by heightened activation of multiple signaling pathways, including the JAK/STAT and PI3K/AKT/mTOR pathways. We hypothesized that the combined inhibition by JAK2 and mTOR inhibitors would induce an additive antileukemia effect in CRLF2-re Ph-like B-ALL. In this study, we tested the antileukemia efficacy of the type I JAK inhibitor ruxolitinib and type II JAK inhibitor NVP-BBT594 (hereafter abbreviated BBT594) [1] alone and combined with allosteric mTOR inhibitor rapamycin and a second generation ATP-competitive mTOR kinase inhibitor AZD2014. We found that BBT594/AZD2014 combination produced robust anti-leukemic effects in Ph-like cell lines in vitro and in patient-derived xenograft (PDX) cells cultured ex vivo. JAK2/mTOR inhibition arrested the cell cycle and reduced cell survival to a greater extent in Ph-like B-ALL cells with CRLF2-re and JAK2 mutation. Synergistic cell killing was associated with the greater inhibition of JAK2 phosphorylation by BBT594 than by ruxolitinib and the greater inhibition of AKT and 4E-BP1 phosphorylation by AZD2014 than by rapamycin. In vivo, BBT594/AZD2014 co-treatment was most efficacious in reducing spleen size in three Ph-like PDX models, and markedly depleted bone marrow and spleen ALL cells in an ATF7IP-JAK2 fusion PDX. In summary, combined inhibition of JAK/STAT and mTOR pathways by next-generation inhibitors had promising antileukemia efficacy in preclinical models of CRFL2-re Ph-like B-ALL.


Frontiers in Oncology | 2018

Initial Report of a Phase I Study of LY2510924, Idarubicin, and Cytarabine in Relapsed/Refractory Acute Myeloid Leukemia

Prajwal Boddu; Gautam Borthakur; Mythili Koneru; Xuelin Huang; Kiran Naqvi; William G. Wierda; Prithviraj Bose; Elias Jabbour; Zeev Estrov; Jan A. Burger; Yesid Alvarado; April Deshmukh; Ami Patel; Antonio Cavazos; Lina Han; Jorge Cortes; Hagop M. Kantarjian; Michael Andreeff; Marina Konopleva

Background: The CXCR4/SDF-1α axis plays a vital role in the retention of stem cells within the bone marrow and downstream activation of cell survival signaling pathways. LY2510924, a second generation CXCR4, showed significant anti-leukemia activity in a murine AML model. Methods: We conducted a phase I study to determine the safety and toxicity of LY2510924, idarubicin and cytarabine (IA) combination therapy in relapsed/refractory (R/R) AML. Eligible patients were 18–70 years of age receiving up to salvage 3 therapy. A peripheral blood absolute blast count of < 20,000/μL was required for inclusion. LY2510924 was administered daily for 7 days followed by IA from day 8. Two dose escalation levels (10 and 20 mg) were evaluated, with a plan to enroll up to 12 patients in the phase I portion. Results: The median age of the enrolled patients (n = 11) was 55 years (range, 19–70). Median number of prior therapies was 1 (1–3). Six and five patients were treated at dose-levels “0” (10 mg) and “1” (20 mg), respectively. Only one patient experiencing a dose limiting toxicity (grade 3 rash and myelosuppression). Three and one complete responses were observed at dose-levels “0” and “1,” respectively; the overall response rate (ORR) was 36% (4 of 11 patients). A ≥ 50% decrease in CXCR4 mean fluorescence intensity was observed in 4 of 9 patients by flow cytometry, indicating incomplete suppression of CXCR4-receptor occupancy. Conclusions: The combination of LY2510924 with IA is safe in R/R AML. Dose-escalation to a 30 mg LY2510924 dose is planned to achieve complete blockade of CXCR4 receptor occupancy, followed by expansion phase at the recommended phase 2 dose-level.


Blood | 2016

Pre-Clinical Studies of Anti-CD123 CAR-T Cells for the Treatment of Blastic Plasmacytoid Dendritic Cell Neoplasm (BPDCN)

Tianyu Cai; Roman Galetto; Agnès Gouble; Julianne Smith; Antonio Cavazos; Sergej Konoplev; Andrew A. Lane; Monica L. Guzman; Hagop M. Kantarjian; Naveen Pemmaraju; Marina Konopleva


Blood | 2016

Targeting MAPK Signaling Pathway with Cobimetinib (GDC-0973) Enhances Anti-Leukemia Efficacy of Venetoclax (ABT-199/GDC-0199) in Acute Myeloid Leukemia Models

Lina Han; Qi Zhang; Ce Shi; Antonio Cavazos; Vivian Ruvolo; Joel D. Leverson; Dail Monique; Darren C. Phillips; Jun Chen; Sha S Jin; Rodrigo Jacamo; Naval Daver; Elias Jabbour; Hagop M. Kantarjian; Michael Andreeff; Deepak Sampath; Marina Konopleva


Journal of Clinical Oncology | 2018

Initial report of a phase I study of LY2510924 with idarubicin and cytarabine (IA) in relapsed/refractory (R/R) AML.

Prajwal Boddu; Gautam Borthakur; Kiran Naqvi; William G. Wierda; Prithviraj Bose; Elias Jabbour; Zeev Estrov; Jan A. Burger; Yesid Alvarado; April Deshmukh; Ami Patel; Antonio Cavazos; Lina Han; Hagop M. Kantarjian; Michael Andreeff; Marina Konopleva


Cancer Research | 2018

Abstract LB-261: Targeting BET Family Bromodomain with ABBV-075 and BCL-2 with venetoclax (ABT-199) is synergistic in primary acute myeloid leukemia models

Tianyu Cai; Vinitha Mary Kuruvilla; Tamar Uziel; Qi Zhang; Lina Han; Antonio Cavazos; Yu Shen; Marina Konopleva


Cancer Research | 2018

Abstract 1875: Oxidative metabolism as a novel therapeutic target to eradicate T-ALL with mitochondrial complex I inhibitor IACS-010759

Natalia Baran; Alessia Lodi; Shannon R. Sweeney; Vinitha Mary Kuruvilla; Antonio Cavazos; Anna Skwarska; Sriram Shanmuga Velandy; Karine Harutyunyan; Ningping Feng; Marcin Kaminski; Elias Jabbour; Adolfo A. Ferrando; M. Emilia Di Francesco; Joseph R. Marszalek; Stefano Tiziani; Marina Konopleva


Cancer Research | 2018

Abstract 2560: Preclinical efficacy of allogeneic anti-CD123 CAR T-cells for the therapy of blastic plasmacytoid dendritic cell neoplasm (BPDCN)

Tianyu Cai; Kathryn L. Black; Ammar S. Naqvi; Roman Galetto; Agnès Gouble; Julianne Smith; Antonio Cavazos; Lina Han; Qi Zhang; Vinitha Mary Kuruvilla; Sergej Konoplev; Sattva S. Neelapu; Andrew A. Lane; Monica L. Guzman; Hagop M. Kantarjian; Andrei Thomas-Tikhonenko; Naveen Pemmaraju; Marina Konopleva


Clinical Lymphoma, Myeloma & Leukemia | 2017

Targeting the Mitochondria Metabolism of T-ALL with Novel Mitochondrial Complex I Inhibitor IACS-010759

Natalia Baran; Alessia Lodi; Shannon R. Sweeney; Vinitha Mary Kuruvilla; Antonio Cavazos; Sriram Shanmuga Velandy; Karine Harutyunyan; Ningping Feng; Marcin Kaminski; Elias Jabbour; Adolfo A. Ferrando; M. Emilia Di Francesco; Stefano Tiziani; Joseph R. Marszalek; Marina Konopleva

Collaboration


Dive into the Antonio Cavazos's collaboration.

Top Co-Authors

Avatar

Marina Konopleva

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lina Han

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elias Jabbour

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hagop M. Kantarjian

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Qi Zhang

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Michael Andreeff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tianyu Cai

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Vinitha Mary Kuruvilla

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alessia Lodi

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge