Network


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

Hotspot


Dive into the research topics where Mary Beth Rios is active.

Publication


Featured researches published by Mary Beth Rios.


Leukemia | 2006

Frequency and clinical significance of BCR-ABL mutations in patients with chronic myeloid leukemia treated with imatinib mesylate

Elias Jabbour; H. Kantarjian; Dan Jones; Moshe Talpaz; N. Bekele; Stephen J. O'Brien; Xian Zhou; Rajyalakshmi Luthra; Guillermo Garcia-Manero; Francis J. Giles; Mary Beth Rios; Srdan Verstovsek; Jorge Cortes

Mutations of the BCR-ABL kinase domain are a common mechanism of resistance to imatinib in chronic myeloid leukemia. We screened for mutations 171 patients failing imatinib therapy. Sixty-six mutations in 23 amino acids were identified in 62 (36%) patients not responding to imatinib. Phosphate-binding loop (P-loop) mutations were the most frequent (n=24; 36%). By multivariate analysis, factors associated with development of mutations were older age (P=0.026) prior interferon therapy (P=0.026), and accelerated phase or blast phase at time of imatinib failure (P=0.001). After a median follow-up of 38 months (range, 4–68 months) from the start of imatinib therapy, seven patients with non-P-loop and two with P-loop mutation died. By multivariate analysis, development of clonal evolution and higher percentage of peripheral blood basophils were associated with worse survival from the time of imatinib failure. Mutation status had no impact on survival. When survival was measured from the time therapy started, non-P-loop mutations together with duration of response and transformation at the time of failure to imatinib were associated with shorter survival. In conclusion, P-loop mutations were not associated with poor outcome, suggesting that the prognosis of patients who fail imatinib is multifactorial.


Clinical Cancer Research | 2005

Molecular Responses in Patients with Chronic Myelogenous Leukemia in Chronic Phase Treated with Imatinib Mesylate

Jorge Cortes; Moshe Talpaz; Susan O'Brien; Dan Jones; Rajyalakshmi Luthra; Jenny Shan; Francis J. Giles; Stefan Faderl; Srdan Verstovsek; Guillermo Garcia-Manero; Mary Beth Rios; Hagop M. Kantarjian

Purpose: To determine the clinical significance of molecular response and relapse among patients with chronic myelogenous leukemia (CML) treated with imatinib. Experimental Design: We analyzed the results of quantitative PCR in 280 patients with CML in chronic phase who achieved complete cytogenetic remission with imatinib (117 after IFN-α failure and 163 previously untreated). Median follow-up was 31 months (range, 3-52 months). Results: Median BCR-ABL/ABL ratio before the start of therapy was 39.44 (range, 0.252-170.53). A major molecular response (BCR-ABL/ABL ratio <0.05%) was achieved in 174 (62%), and transcripts became undetectable (complete molecular response) in 95 (34%). By multivariate analysis, only treatment with high-dose imatinib (P = 0.02) was associated with achievement of a major molecular response. Nine of 166 (5%) patients who achieved a major molecular response lost their cytogenetic remission, compared with 25 of 68 (37%) among those who did not achieve this response (P < 0.0001). Patients achieving a major molecular response 12 months after the start of therapy had significantly better complete cytogenetic remission duration than others. A >1-log reduction in transcript levels after 3 months of therapy predicted for an improved probability of achieving a major molecular response at 24 months. Increasing levels of BCR-ABL transcripts predicted for a loss of cytogenetic remission only among patients who did not achieve a major molecular response. Conclusions: Achieving a major molecular response, particularly within the first year of therapy, is predictive of a durable cytogenetic remission and may be the future goal of therapy in CML.


Lancet Oncology | 2012

Inotuzumab ozogamicin, an anti-CD22–calecheamicin conjugate, for refractory and relapsed acute lymphocytic leukaemia: a phase 2 study

Hagop M. Kantarjian; Deborah A. Thomas; Jeffrey L. Jorgensen; Elias Jabbour; Partow Kebriaei; Michael Rytting; Sergernne York; Farhad Ravandi; Monica Kwari; Stefan Faderl; Mary Beth Rios; Jorge Cortes; Luis Fayad; Robert Tarnai; Sa A. Wang; Richard E. Champlin; Anjali S. Advani; Susan O'Brien

BACKGROUND The outlook for patients with refractory and relapsed acute lymphocytic leukaemia (ALL) is poor. CD22 is highly expressed in patients with ALL. Inotuzumab ozogamicin is a CD22 monoclonal antibody conjugated to the toxin calecheamicin. We did a phase 2 study to assess the efficacy of this antibody. METHODS We recruited patients at the MD Anderson Cancer Center, Houston, TX, USA, between June, 2010, and March, 2011. Adults and children with refractory and relapsed ALL were eligible. Ten adults were treated before enrolment of children started. Patients were given 1·8 mg/m(2) inotuzumab ozogamicin intravenously over 1 h every 3-4 weeks (the first three adults and three children received 1·3 mg/m(2) in the first course). The primary endpoint was overall response (complete response or marrow complete response with no recovery of platelet count or incomplete recovery of neutrophil and platelet counts). Analysis was done by intention to treat. This study is registered, number NCT01134575. FINDINGS 49 patients were enrolled and treated. Median age was 36 years (range 6-80). CD22 was expressed in more than 50% of blasts in all patients. The median number of courses was two (range one to five) and the median time between courses was 3 weeks (range 3-6). Nine (18%) patients had complete response, 19 (39%) had marrow complete response, 19 (39%) had resistant disease, and two (4%) died within 4 weeks of starting treatment. The overall response rate was 57% (95% CI 42-71). The most frequent adverse events during course one of treatment were fever (grade 1-2 in 20 patients, grade 3-4 in nine), hypotension (grade 1-2 in 12 patients, grade 3 in one), and liver-related toxic effects (bilirubin: grade 1-2 in 12 patients, grade 3 in two; raised aminotransferase concentration: grade 1-2 in 27 patients, grade 3 in one). INTERPRETATION Inotuzumab ozogamicin shows promise as a treatment for refractory and relapsed ALL. FUNDING Pfizer.


Cancer | 2003

Results of decitabine (5-aza-2'deoxycytidine) therapy in 130 patients with chronic myelogenous leukemia.

Hagop M. Kantarjian; Susan O'Brien; Jorge Cortes; Francis J. Giles; Stefan Faderl; Jean-Pierre Issa; Guillermo Garcia-Manero; Mary Beth Rios; Jianqin Shan; Michael Andreeff; Michael J. Keating; Moshe Talpaz

General and site‐specific DNA methylation is associated with tumor progression and resistance in several cancers, including chronic myelogenous leukemia (CML). Decitabine is a hypomethylating agent that has shown encouraging preliminary anti‐CML activity. This study evaluated the activity and toxicity of decitabine in different phases of CML.


Cancer | 1999

Chronic myelogenous leukemia in nonlymphoid blastic phase: Analysis of the results of first salvage therapy with three different treatment approaches for 162 patients

Stefano Sacchi; Hagop Kantarjian; Susan O'Brien; Jorge Cortes; Mary Beth Rios; Francis J. Giles; M. Beran; Charles A. Koller; Michael J. Keating; Moshe Talpaz

The prognoses of patients with chronic myelogenous leukemia in blastic phase (CML‐BP) are extremely poor. Treatment of patients with nonlymphoid CML‐BP is associated with very low response rates, a median survival of 2–3 months, and significant toxicities. The aim of this study was to evaluate the results of therapy in CML‐BP with different treatments in relation to response rate, survival, and toxicity.


Journal of Clinical Oncology | 2003

Phase I Clinical and Pharmacology Study of Clofarabine in Patients With Solid and Hematologic Cancers

Hagop M. Kantarjian; Varsha Gandhi; Peter Kozuch; Stefan Faderl; Francis J. Giles; Jorge Cortes; Susan O'Brien; Nuhad K. Ibrahim; Fadlo R. Khuri; Min Du; Mary Beth Rios; Sima Jeha; Peter McLaughlin; William Plunkett; Michael J. Keating

PURPOSE To define the maximum-tolerated doses (MTDs) and dose-limiting toxicities (DLTs) of clofarabine, given as a 1-hour infusion daily for 5 days, in patients with solid tumors and with acute leukemia. PATIENTS AND METHODS The initial part of the study defined the MTD and DLT in solid tumors. The second part of the study defined the MTD and DLT in acute leukemia. RESULTS The starting dose of clofarabine (15 mg/m(2)) was myelosuppressive, requiring several dose de-escalations to 2 mg/m(2), the dose suggested for phase II studies in solid tumors. Dose escalation in acute leukemia started at 7.5 mg/m(2), with several escalations to 55 mg/m(2). The DLT was reversible hepatotoxicity at 55 mg/m(2). The recommended dose for acute leukemia phase II studies was 40 mg/m(2). Among 32 treated patients with acute leukemia, two achieved a complete response and three had a marrow complete response without platelet recovery (hematologic improvement), for an overall response rate of 16%. At 40 mg/m(2), the median plasma clofarabine level was 1.5 micro mol/L (range, 0.42 to 3.2 micro mol/L; n = 7). Cellular and plasma pharmacokinetic studies suggested dose proportionality but showed a wide variation in intracellular concentrations of clofarabine triphosphate. CONCLUSION This phase I study defined the following two MTDs for clofarabine given as a 1-hour infusion daily for 5 days: 2 mg/m(2) for solid tumors, the DLT being myelosuppression; and 40 mg/m(2) for acute leukemia, the DLT being hepatotoxicity. Encouraging activity was observed in acute leukemia.


Blood | 2012

Improved survival in chronic myeloid leukemia since the introduction of imatinib therapy: a single-institution historical experience

Hagop M. Kantarjian; Susan O'Brien; Elias Jabbour; Guillermo Garcia-Manero; Alfonso Quintás-Cardama; Jenny Shan; Mary Beth Rios; Farhad Ravandi; Stefan Faderl; Tapan Kadia; Gautam Borthakur; Xuelin Huang; Richard E. Champlin; Moshe Talpaz; Jorge Cortes

A total of 1569 patients with chronic myeloid leukemia (CML) referred to our institution within 1 month of diagnosis since 1965 were reviewed: 1148 chronic phase (CP), 175 accelerated phase (AP), and 246 blastic phase (BP). The median survival was 8.9 years in CP, 4.8 years in AP, and 6 months in BP. In CP, the 8-year survival was ≤ 15% before 1983, 42%-65% from 1983-2000, and 87% since 2001. Survival was worse in older patients (P = .004), but this was less significant since 2001 (P = .07). Survival by Sokal risk was significantly different before 2001 (P < .001), but not since 2001 (P = .4). In AP, survival improved over time (P < .001); the 8-year survival in patients treated since 2001 was 75%. Survival by age was not different in years < 2001 (P = .09), but was better since 2001 in patients ≤ 70 years of age (P = .004). In BP, the median survival improved over time (P < .001), although it has been only 7 months since 2001. In summary, survival in CML has significantly improved since 2001, particularly so in CP-AML and AP-CML. Imatinib therapy minimized the impact of known prognostic factors and Sokal risk in CP-CML and accentuated the impact of age in AP- and BP-CML.


Journal of Clinical Oncology | 2006

Pregnancy Among Patients With Chronic Myeloid Leukemia Treated With Imatinib

Patricia Ault; Hagop M. Kantarjian; Susan O'Brien; Stefan Faderl; Miloslav Beran; Mary Beth Rios; Charles Koller; Francis J. Giles; Michael J. Keating; Moshe Talpaz; Jorge Cortes

PURPOSE Imatinib has potential teratogenicity in animals, but the effect of exposure to imatinib during conception and pregnancy in humans is not known. PATIENTS AND METHODS The records of all patients with chronic myeloid leukemia (CML) treated with imatinib were reviewed. We report the experience on 19 pregnancies involving 18 patients (10 females and eight males) who conceived while receiving imatinib for the treatment of CML. RESULTS All female patients discontinued therapy immediately on recognition of pregnancy. Three pregnancies (involving two female patients and one male patient) ended in spontaneous abortion, and one patient had an elective abortion. All other pregnancies were uneventful. Two of the 16 babies had minor abnormalities at or shortly after birth (hypospadias in one baby and rotation of small intestine in one baby) that were surgically repaired. All babies have continued normal growth and development. Among female patients who interrupted therapy, five of nine in complete hematologic remission (CHR) at the time of treatment interruption eventually lost CHR, and six experienced an increase in Philadelphia chromosome-positive metaphases. At a median of 18 months after resuming therapy with imatinib, eight patients had a cytogenetic response (complete in three patients). CONCLUSION Although there is no evidence that a brief exposure to imatinib during conception and pregnancy adversely affects the developing fetus, most patients lose their response after treatment interruption. Patients receiving imatinib should be advised to practice adequate contraception.


Molecular Cancer Therapeutics | 2008

CXCR4 up-regulation by imatinib induces chronic myelogenous leukemia (CML) cell migration to bone marrow stroma and promotes survival of quiescent CML cells

Linhua Jin; Yoko Tabe; Sergej Konoplev; Yuanyuan Xu; Clinton E. Leysath; Hongbo Lu; Shinya Kimura; Akimichi Ohsaka; Mary Beth Rios; Leslie Calvert; Hagop M. Kantarjian; Michael Andreeff; Marina Konopleva

Chronic myelogenous leukemia (CML) is driven by constitutively activated Bcr-Abl tyrosine kinase, which causes the defective adhesion of CML cells to bone marrow stroma. The overexpression of p210Bcr-Abl was reported to down-regulate CXCR4 expression, and this is associated with the cell migration defects in CML. We proposed that tyrosine kinase inhibitors, imatinib or INNO-406, may restore CXCR4 expression and cause the migration of CML cells to bone marrow microenvironment niches, which in turn results in acquisition of stroma-mediated chemoresistance of CML progenitor cells. In KBM5 and K562 cells, imatinib, INNO-406, or IFN-α increased CXCR4 expression and migration. This increase in CXCR4 levels on CML progenitor cells was likewise found in samples from CML patients treated with imatinib or IFN-α. Imatinib induced G0-G1 cell cycle block in CML cells, which was further enhanced in a mesenchymal stem cell (MSC) coculture system. MSC coculture protected KBM-5 cells from imatinib-induced cell death. These antiapoptotic effects were abrogated by the CXCR4 antagonist AMD3465 or by inhibitor of integrin-linked kinase QLT0267. Altogether, these findings suggest that the up-regulation of CXCR4 by imatinib promotes migration of CML cells to bone marrow stroma, causing the G0-G1 cell cycle arrest and hence ensuring the survival of quiescent CML progenitor cells. [Mol Cancer Ther 2008;7(1):48–58]


Leukemia & Lymphoma | 1993

Fludarabine and Arabinosylcytosine Therapy of Refractory and Relapsed Acute Myelogenous Leukemia

Elihu H. Estey; William Plunkett; Varsha Gandhi; Mary Beth Rios; Hagop M. Kantarjian; Michael J. Keating

There is a strong association between ability of leukemia blasts to accumulate ara-CTP, the active metabolite of ara-C, and response to ara-C in patients with relapsed or refractory AML. Ara-C dose rates in excess of 0.5 g/m2/h do not produce further ara-CTP formation. In contrast, when given 4 h prior to ara-C at this dose rate, fludarabine, at doses that are free of neurotoxicity in CLL, enhances ara-CTP accumulation. This led us to administer fludarabine and ara-C to 59 patients with AML in relapse or unresponsive to initial therapy. Fludarabine was given at 30 mg/m2 once daily for 5 doses and ara-C at 0.5 g/m2/h for 2-6 h daily for 6 doses. Doses of fludarabine preceded those of ara-C by 4 h. Results with fludarabine and ara-C (FA) were compared with those of patients treated at M.D. Anderson with high-dose ara-C (HDAC) or intermediate-dose ara-C (IDAC). The complete remission rate with FA was 21/59 (36%) and the actuarial median CR duration 39 weeks. FA produced significantly higher remission rates than HDAC or IDAC in patients with initial remissions > 1 yr (14/20 vs 9/23 vs 6/18, p < 0.05). Response rates were similar for all three treatments in patients with initial remissions < 1 yr or with primary refractory disease. The regimen was well tolerated; one patient developed peripheral neuropathy. This low level of toxicity encourages combination with other antileukemia agents.

Collaboration


Dive into the Mary Beth Rios's collaboration.

Top Co-Authors

Avatar

Hagop M. Kantarjian

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jorge Cortes

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar

Susan O'Brien

University of California

View shared research outputs
Top Co-Authors

Avatar

Moshe Talpaz

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Guillermo Garcia-Manero

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Stefan Faderl

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Francis J. Giles

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar

Srdan Verstovsek

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

Jianqin Shan

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge