Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Transplant Cell Ther ; 2024 May 18.
Article in English | MEDLINE | ID: mdl-38768906

ABSTRACT

Multiplexed gastrointestinal PCR panels (MGPPs) are frequently used to aid the diagnosis and management of diarrhea in hematopoietic stem cell transplantation (HCT) recipients. Many issues related to the optimal use of MGPPs in HCT patients remain to be clarified. We aimed to better define MGPP diagnostic and therapeutic stewardship in HCT recipients, including indications for and benefits of testing, optimal timing of tests, and interpretation of results. We retrieved 463 consecutive MGPPs ordered on 651 consecutive first HCT (312 allogeneic, 339 autologous) performed at our institution between June 2015 and June 2023. One hundred and sixteen of the 463 MGPPs (25%) identified at least 1 diarrheagenic pathogen, and 12 (3%) identified more than 1 diarrheagenic pathogen. A positive result was more likely if the test was ordered within 48 hours of a hospital admission (41%; 32 of 78) or as an outpatient (41%; 46 of 111) compared with evaluation of hospital-onset diarrhea (14%; 38 of 274). Among the positive results, the most frequent pathogens identified included Clostridioides difficile (64%), diarrheagenic Escherichia coli (20%), norovirus (9%), and adenovirus 40/41 (5%). Thirty-eight percent of the positive C. difficile MGPP determinations were associated with a positive test for toxin. In our allogeneic HCT cohort, 3% of MGPPs for hospital-onset diarrhea yielded an organism other than C. difficile. Fifty-six percent of positive and 14% of all submitted tests resulted in a change in treatment. For organisms other than C. difficile, only 1% of all tests and 5% of positive tests resulted in initiation of therapy. For patients at risk for acute graft-versus-host disease (aGVHD), a positive or negative MGPP result was not predictive of a new diagnosis of aGVHD in proximity to diarrhea onset. These results suggest that MGPP testing is most useful when performed at hospital admission or on an outpatient basis. Because MGPPs are sensitive and do not distinguish between colonization and causes of diarrhea, caution is needed when interpreting results, especially for toxin-negative C. difficile and diarrheagenic gram-negative organisms.

2.
Transplant Cell Ther ; 30(4): 438.e1-438.e11, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38281591

ABSTRACT

The optimal timing of immunosuppression and post-transplantation cyclophosphamide (PTCy) in haploidentical hematopoietic stem cell transplantation (haplo-HSCT) is unknown. However, cytokine release syndrome (CRS) following haplo-HSCT is associated with worse transplantation outcomes, and the incidence of CRS may be affected by the timing of immunosuppression and PTCy. In this study, we compared CRS and other transplantation outcomes in 2 cohorts receiving different immunosuppression and PTCy schedules following haplo-HSCT. This was a retrospective cohort study of 91 patients who underwent haplo-HSCT at the Intermountain Health Blood and Marrow Transplant Program. The original or standard haplo-HSCT GVHD prophylaxis regimen included PTCy on days +3 and +4, with mycophenolate mofetil (MMF) and tacrolimus starting on day +5. The modified regimen adopted in November 2020 changed PTCy to days +3 and +5, with earlier introduction of tacrolimus and MMF, on day -1 and day 0, respectively. Grade ≥1 CRS occurred in 32% of patients in the modified regimen, in 82% of patients in the standard regimen (P <.0001), and 65% overall. Likewise, grade ≥2 CRS was lower with the modified regimen (16% versus 57%; P = .0002). The mean duration of CRS symptoms was longer with the standard regimen (3.14 days versus 1.44 days; P = .0003). The incidence of acute graft-versus-host disease grade III-IV or extensive chronic GVHD (cGVHD) at 1 year was lower in the modified regimen (6% versus 32%; P = .0068). No differences between the standard and modified regimens were seen in overall survival, relapse, or GVHD-free relapse-free survival (GRFS), although there appeared to be a trend toward improved GRFS with the modified regimen. Post hoc analysis comparing GRFS in patients with CRS and those without CRS found that CRS was associated with lower GRFS at 1 year (36% versus 63%; P = .0138). The duration of broad-spectrum antibiotic therapy was decreased by 7.5 days (P = .0017) and the time to hospital discharge was reduced by 7.1 days (P = .0241) with the modified regimen. This is the first analysis to evaluate and find a difference in CRS with early initiation of immunosuppressive therapy in haplo-HSCT. Our results suggest that this modified GVHD regimen benefits patients by reducing CRS and high-grade GVHD compared to the standard PTCy-based GVHD prophylaxis regimen in haplo-HSCT. Additionally, this novel regimen did not appear to negatively impact outcomes.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Tacrolimus/therapeutic use , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/drug therapy , Retrospective Studies , Transplantation Conditioning/methods , Neoplasm Recurrence, Local/complications , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Cyclophosphamide/therapeutic use , Graft vs Host Disease/epidemiology , Graft vs Host Disease/prevention & control , Mycophenolic Acid/therapeutic use , Immunosuppression Therapy/adverse effects
3.
Transplant Cell Ther ; 29(7): 440-448, 2023 07.
Article in English | MEDLINE | ID: mdl-37031747

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is the most prevalent subtype of non-Hodgkin lymphoma. Although outcomes to frontline therapy are encouraging, patients who are refractory to or in relapse after first-line therapy experience inferior outcomes. A significant proportion of patients treated with additional lines of cytotoxic chemotherapy ultimately succumb to their disease, as established in the SCHOLAR-1 study. Chimeric antigen receptor (CAR) T cell therapy is a novel approach to cancer management that reprograms a patient's own T cells to better target and eliminate cancer cells. It was initially approved by the US Food and Drug Administration for patients with relapsed/refractory (r/r) DLBCL as a third line of treatment. Based on recently published randomized data, CAR-T therapy (axicabtagene ciloleucel and lisocabtagene maraleucel) also has been approved as a second line of treatment for patients who are primary refractory or relapse within 12 months of first-line therapy. Despite the proven efficacy in treating r/r DLBCL with CD19-directed CAR-T therapy, several barriers may prevent eligible patients from receiving treatment. Barriers to CAR-T therapy include cost of therapy, patient hesitancy, required travel to academic treatment centers, nonreferrals, poor understanding of CAR-T therapy, lack of caregiver support, knowledge of available resources, and timely patient selection by referring oncologists. In this review, we provide an overview of the FDA-approved CD19-directed CAR-T cell therapies (tisagenlecleucel, axicabtagene ciloleucel, and lisocabtagene maraleucel) from pivotal clinical trials and supporting real-world evidence from retrospective studies. In both clinical trials and real-world settings, CAR-T therapy has been shown to be safe and efficacious for treating patients with r/r DLBCL: however, several barriers prevent eligible patients from accessing these therapies. Barriers to referrals for CAR-T therapy are described, along with recommendations to improve collaboration between community oncologists and physicians from CAR-T therapy treatment centers and subsequent long-term care of patients in community treatment centers.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Lymphoma, Non-Hodgkin , Receptors, Chimeric Antigen , United States , Humans , Receptors, Chimeric Antigen/therapeutic use , Retrospective Studies , Neoplasm Recurrence, Local/drug therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Non-Hodgkin/chemically induced , Lymphoma, Non-Hodgkin/drug therapy , Antigens, CD19/therapeutic use , Referral and Consultation , Cell- and Tissue-Based Therapy
4.
Transplant Cell Ther ; 29(1): 54.e1-54.e6, 2023 01.
Article in English | MEDLINE | ID: mdl-36208727

ABSTRACT

The mechanism(s) of acquisition of extended-spectrum cephalosporin-resistant Enterobacteriaceae (ESCRE) on inpatient hospital units dedicated to hematopoietic stem cell transplantation (HSCT) is unclear. The objectives of this study were to determine whether ESCRE organisms are transmitted among patients housed on a HSCT unit, clarify the mechanisms involved, and determine whether routine surveillance for ESCRE carriage and contact isolation for ESCRE carriers is beneficial. The study was conducted on a 30-bed inpatient unit dedicated to the care of patients with hematologic malignancies and HSCT recipients. To investigate whether ESCRE organisms may be transmitted vertically to subsequent room occupants, presumably through contamination of room surfaces, we (1) cultured 6 high touch areas in 10 rooms before and 9 rooms after terminal cleaning that had been occupied by patients with ESCRE carriage, (2) determined the in vitro survivals of our most common clinical ESCRE species, and (3) followed the subsequent room occupants of 54 consecutive ESCRE colonized patients for the development of inpatient acquired ESCRE carriage. To investigate whether ESCRE organisms are transmitted horizontally among inpatients we (1) sequenced 60 available ESCRE Escherichia coli isolates obtained from unit inpatients and searched for identities using complete-genome multisequence locus typing (cgMLST) and (2) retrospectively tabulated the cumulative rates of acquired ESCRE carriage in 356 patients admitted for a first HSCT before (200 patients) or after (156 patients) institution of universal ESCRE stool surveillance and contact isolation for carriers. No ESCRE organisms were cultured from patient rooms before or after terminal cleaning. In vitro, few, if any, ESCRE organisms survived longer than 2 hours. Nine of the subsequent occupants of a room in which a patient with ESCRE carriage had resided were detected with ESCRE carriage, only 2 of whom carried the same species as that of the prior occupant. DNA sequencing and cgMLST determination of the 60 E. coli isolates showed 53 cgMLST strains. Seven of the 53 strains were shared by 2 patients. After institution of universal ESCRE surveillance/isolation there was a significant decline in acquired ESCRE carriage among HSCT recipients. We conclude that vertical transmission of ESCRE organisms through room contamination appears to be uncommon on modern HSCT units. Conversely, our results are consistent with the horizontal spread of ESCRE organisms, probably mediated by intermediate vectors such as personnel or shared equipment. Further studies are needed to better define the magnitude of and risk factors for ESCRE horizontal transfers and the benefits of ESCRE surveillance/isolation.


Subject(s)
Cross Infection , Hematopoietic Stem Cell Transplantation , Humans , Enterobacteriaceae/genetics , Enterobacteriaceae/metabolism , Cephalosporins/therapeutic use , Cephalosporins/metabolism , Escherichia coli/metabolism , Retrospective Studies , Cross Infection/prevention & control , beta-Lactamases/genetics , beta-Lactamases/metabolism , Monobactams/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects
5.
Future Oncol ; 18(19): 2415-2431, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35583358

ABSTRACT

Background: Chimeric antigen receptor T-cell (CAR-T) therapy represents a new frontier in multiple myeloma. It is important to understand critical success factors (CSFs) that may optimize its use in this therapeutic area. Methods: We estimated the CAR-T process using time-driven activity-based costing. Information was obtained through interviews at four US oncology centers and with payer representatives, and through publicly available data. Results: The CAR-T process comprises 13 steps which take 177 days; it was estimated to include 46 professionals and ten care settings. CSFs included proactive collaboration, streamlined reimbursement and CAR-T administration in alternative settings when possible. Implementing CSFs may reduce episode time and costs by 14.4 and 13.2%, respectively. Conclusion: Our research provides a blueprint for improving efficiencies in CAR-T therapy, thereby increasing its sustainability for multiple myeloma.


Patients with multiple myeloma can now be treated with chimeric antigen receptor T-cell (CAR-T) therapy. We studied how CAR-T therapy is used for multiple myeloma. We also studied things that could help make this therapy easier for doctors to use. The CAR-T process takes 13 steps and 177 days. It begins with the choice to use the therapy and ends about 100 days after it is used. The process uses 46 different healthcare professionals and ten different locations. We found several possible changes that can improve this process. Of these changes, three stand out. First, improved teamwork between members of the care team can help them prepare for and resolve possible problems. Second, reducing insurance red tape will make it easier to provide CAR-T therapy to patients. Third, allowing use of CAR-T therapy in places other than hospitals can help more patients receive this therapy. If applied, these three things may lower the time needed to treat patients by 14.4% and may reduce costs by 13.2%.


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
6.
EClinicalMedicine ; 47: 101409, 2022 May.
Article in English | MEDLINE | ID: mdl-35475258

ABSTRACT

Background: In COVACTA, a randomised, placebo-controlled trial in patients hospitalised with coronavirus disease-19 (COVID-19), tocilizumab did not improve 28-day mortality, but shortened hospital and intensive care unit stay. Longer-term effects of tocilizumab in patients with COVID-19 are unknown. Therefore, the efficacy and safety of tocilizumab in COVID-19 beyond day 28 and its impact on Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) clearance and antibody response in COVACTA were investigated. Methods: Adults in Europe and North America hospitalised with COVID-19 (N = 452) between April 3, 2020 and May 28, 2020 were randomly assigned (2:1) to double-blind intravenous tocilizumab or placebo and assessed for efficacy and safety through day 60. Assessments included mortality, time to hospital discharge, SARS-CoV-2 viral load in nasopharyngeal swab and serum samples, and neutralising anti-SARS-CoV-2 antibodies in serum. ClinicalTrials.gov registration: NCT04320615. Findings: By day 60, 24·5% (72/294) of patients in the tocilizumab arm and 25·0% (36/144) in the placebo arm died (weighted difference -0·5% [95% CI -9·1 to 8·0]), and 67·0% (197/294) in the tocilizumab arm and 63·9% (92/144) in the placebo arm were discharged from the hospital. Serious infections occurred in 24·1% (71/295) of patients in the tocilizumab arm and 29·4% (42/143) in the placebo arm. Median time to negative reverse transcriptase-quantitative polymerase chain reaction result in nasopharyngeal/oropharyngeal samples was 15·0 days (95% CI 14·0 to 21·0) in the tocilizumab arm and 21·0 days (95% CI 14·0 to 28·0) in the placebo arm. All tested patients had positive test results for neutralising anti-SARS-CoV-2 antibodies at day 60. Interpretation: There was no mortality benefit with tocilizumab through day 60. Tocilizumab did not impair viral clearance or host immune response, and no new safety signals were observed. Future investigations may explore potential biomarkers to optimize patient selection for tocilizumab treatment and combination therapy with other treatments. Funding: F. Hoffmann-La Roche Ltd and the US Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, under OT number HHSO100201800036C.

7.
Exp Hematol Oncol ; 11(1): 10, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35227310

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of relapsed/refractory B-cell malignancies. However, there is no data on the safety and efficacy of CAR T-cell therapy in patients with end stage renal disease (ESRD) requiring dialysis. In this report, we present two patients with DLBCL and ESRD who were successfully treated with different CAR T-cell products. Patient #1 is a 66 year-old woman with a history of HIV who was treated to complete response with axicabtagene ciloleucel with treatment complicated by grade 1 cytokine release syndrome (CRS) and grade 2 immune effector cell-associated neurolotoxicity syndrome (ICANS). Patient #2 is 52 year old woman whose ESRD was caused by ifosphamide toxicity and was treated to complete response with lisocabtagene maraleucel and did not experience either CRS or ICANS. Both patients received lymphodepletion chemotherapy with fludarabine and cyclophosphamide, which was dose-adjusted for ESRD with scheduled dialysis 12 h after each dose of lymphodepletion chemotherapy. Patients with DLBCL and ESRD can be safely administered both lymphodepletion chemotherapy and CAR T-cell therapy. Additionally, the fact that both patients achieved complete response to therapy suggests that CAR T-cell therapy should be strongly considered in patients with ESRD. Long-term follow up is needed to determine if therapy in this setting is of curative intent.

8.
Intensive Care Med ; 47(11): 1258-1270, 2021 11.
Article in English | MEDLINE | ID: mdl-34609549

ABSTRACT

PURPOSE: Trials of tocilizumab in patients with severe COVID-19 pneumonia have demonstrated mixed results, and the role of tocilizumab in combination with other treatments is uncertain. Here we evaluated whether tocilizumab plus remdesivir provides greater benefit than remdesivir alone in patients with severe COVID-19 pneumonia. METHODS: This randomized, double-blind, placebo-controlled, multicenter trial included patients hospitalized with severe COVID-19 pneumonia requiring > 6 L/min supplemental oxygen. Patients were randomly assigned (2:1 ratio) to receive tocilizumab 8 mg/kg or placebo intravenously plus ≤ 10 days of remdesivir. The primary outcome was time from randomization to hospital discharge or "ready for discharge" (defined as category 1, assessed by the investigator on a 7-category ordinal scale of clinical status) to day 28. Patients were followed for 60 days. RESULTS: Among 649 enrolled patients, 434 were randomly assigned to tocilizumab plus remdesivir and 215 to placebo plus remdesivir. 566 patients (88.2%) received corticosteroids during the trial to day 28. Median time from randomization to hospital discharge or "ready for discharge" was 14 (95% CI 12-15) days with tocilizumab plus remdesivir and 14 (95% CI 11-16) days with placebo plus remdesivir [log-rank P = 0.74; Cox proportional hazards ratio 0.97 (95% CI 0.78-1.19)]. Serious adverse events occurred in 128 (29.8%) tocilizumab plus remdesivir and 72 (33.8%) placebo plus remdesivir patients; 78 (18.2%) and 42 (19.7%) patients, respectively, died by day 28. CONCLUSIONS: Tocilizumab plus remdesivir did not shorten time to hospital discharge or "ready for discharge" to day 28 compared with placebo plus remdesivir in patients with severe COVID-19 pneumonia.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antimetabolites/therapeutic use , Antiviral Agents , COVID-19 Drug Treatment , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/therapeutic use , Humans
10.
N Engl J Med ; 384(16): 1503-1516, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33631066

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (Covid-19) is associated with immune dysregulation and hyperinflammation, including elevated interleukin-6 levels. The use of tocilizumab, a monoclonal antibody against the interleukin-6 receptor, has resulted in better outcomes in patients with severe Covid-19 pneumonia in case reports and retrospective observational cohort studies. Data are needed from randomized, placebo-controlled trials. METHODS: In this phase 3 trial, we randomly assigned patients who were hospitalized with severe Covid-19 pneumonia in a 2:1 ratio receive a single intravenous infusion of tocilizumab (at a dose of 8 mg per kilogram of body weight) or placebo. Approximately one quarter of the participants received a second dose of tocilizumab or placebo 8 to 24 hours after the first dose. The primary outcome was clinical status at day 28 on an ordinal scale ranging from 1 (discharged or ready for discharge) to 7 (death) in the modified intention-to-treat population, which included all the patients who had received at least one dose of tocilizumab or placebo. RESULTS: Of the 452 patients who underwent randomization, 438 (294 in the tocilizumab group and 144 in the placebo group) were included in the primary and secondary analyses. The median value for clinical status on the ordinal scale at day 28 was 1.0 (95% confidence interval [CI], 1.0 to 1.0) in the tocilizumab group and 2.0 (non-ICU hospitalization without supplemental oxygen) (95% CI, 1.0 to 4.0) in the placebo group (between-group difference, -1.0; 95% CI, -2.5 to 0; P = 0.31 by the van Elteren test). In the safety population, serious adverse events occurred in 103 of 295 patients (34.9%) in the tocilizumab group and in 55 of 143 patients (38.5%) in the placebo group. Mortality at day 28 was 19.7% in the tocilizumab group and 19.4% in the placebo group (weighted difference, 0.3 percentage points; 95% CI, -7.6 to 8.2; nominal P = 0.94). CONCLUSIONS: In this randomized trial involving hospitalized patients with severe Covid-19 pneumonia, the use of tocilizumab did not result in significantly better clinical status or lower mortality than placebo at 28 days. (Funded by F. Hoffmann-La Roche and the Department of Health and Human Services; COVACTA ClinicalTrials.gov number, NCT04320615.).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , COVID-19 Drug Treatment , Receptors, Interleukin-6/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , COVID-19/complications , COVID-19/mortality , COVID-19/therapy , Double-Blind Method , Female , Hospital Mortality , Hospitalization , Humans , Infusions, Intravenous , Intensive Care Units , Male , Middle Aged , Respiration, Artificial , Treatment Failure
11.
J Clin Oncol ; 38(27): 3095-3106, 2020 09 20.
Article in English | MEDLINE | ID: mdl-32667831

ABSTRACT

PURPOSE: Axicabtagene ciloleucel (axi-cel) was approved by the Food and Drug Administration for relapsed aggressive B-cell non-Hodgkin lymphoma in part on the basis of durable remission rates of approximately 40% in a clinical trial population. Whether this efficacy, and the rates of toxicity, would be consistent in a postcommercial setting, with relaxed eligibility criteria and bridging therapy, is unknown. This study describes the efficacy and safety correlates and outcomes in this setting. PATIENTS AND METHODS: One hundred twenty-two patients from 7 medical centers in the United States were treated with axi-cel and were included in a modified intent-to-treat (mITT) analysis. Seventy-six patients (62%) were ineligible for the ZUMA-1 trial. Response and toxicity rates, duration of response (DOR), survival, and covariates are described on the basis of the mITT population. Correlative studies on blood and tumor samples were performed to investigate potential biomarkers of response and resistance. RESULTS: Median follow-up was 10.4 months. In the mITT population, the best overall and complete response (CR) rates were 70% and 50%, respectively. Median DOR and progression-free survival (PFS) were 11.0 and 4.5 months in all patients and were not reached (NR) in CR patients. Median overall survival (OS) was NR; 1-year OS was 67% (95% CI, 59% to 77%). Although response rates were similar in the ZUMA-1-eligible and ZUMA-1-ineligible groups (70% v 68%), there was a statistically significant improvement in CR rate (63% v 42%, P = .016), DOR (median, NR v 5.0 months; P = .014), PFS (median, NR v 3.3 months; P = .020), and OS (1-year OS, 89% v 54%; P < .001) in patients who were ZUMA-1 eligible. Rates of grade ≥ 3 cytokine release syndrome and neurotoxicty were 16% and 35%, respectively. CONCLUSION: Axi-cel yields similar rates of overall response and toxicity in commercial and trial settings, although CR rates and DOR were more favorable in patients eligible for ZUMA-1.


Subject(s)
Antigens, CD19/therapeutic use , Lymphoma, Large B-Cell, Diffuse/therapy , Adult , Aged , Antigens, CD19/adverse effects , Antigens, CD19/metabolism , B7-H1 Antigen/metabolism , Biological Products , Biomarkers/blood , C-Reactive Protein/metabolism , Clinical Trials as Topic , Cytokine Release Syndrome/etiology , Ferritins/blood , Humans , Immunotherapy, Adoptive/adverse effects , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Middle Aged , Neoplasm Grading , Neurotoxicity Syndromes/etiology , Patient Selection , Progression-Free Survival , Receptors, Chimeric Antigen/metabolism , Recurrence , Retrospective Studies , Survival Rate , T-Lymphocytes/metabolism , Young Adult
13.
Curr Hematol Malig Rep ; 15(1): 1-8, 2020 02.
Article in English | MEDLINE | ID: mdl-32034660

ABSTRACT

PURPOSE OF REVIEW: This review discusses the current standard of care for incorporation of FLT3 TKIs and HCT into the treatment of FLT3-ITD AML. Additionally, this review provides an approach to the patient with relapsed/refractory disease. RECENT FINDINGS: Over the last decade, the routine use of HCT as consolidative therapy and the development of FLT3 TKIs have significantly improved remission rates and overall survival. The value and challenges of MRD assessment in FLT3 disease are discussed and current mechanisms of relapse are explored, as are the ongoing questions in the field that current clinical trials are seeking to answer. FLT3-ITD mutations are common in acute myeloid leukemia and historically have been associated with a poor prognosis, but with the incorporation of FLT3 TKIs and routine use of allogeneic stem cell transplant as consolidative therapy, outcomes have improved dramatically. Ongoing research seeks to answer how and when to best use current therapies, and how to overcome resistance to FLT3 inhibition.


Subject(s)
Biomarkers, Tumor/genetics , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Mutation , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Drug Resistance, Neoplasm/genetics , Genetic Predisposition to Disease , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Molecular Targeted Therapy , Phenotype , Protein Kinase Inhibitors/adverse effects , Recurrence , Risk Assessment , Risk Factors , Transplantation, Homologous , Treatment Outcome , fms-Like Tyrosine Kinase 3/metabolism
14.
Expert Opin Biol Ther ; 19(11): 1157-1164, 2019 11.
Article in English | MEDLINE | ID: mdl-31342797

ABSTRACT

Introduction: Traditionally, outcomes for patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma have been poor. There has been a clear need for effective therapeutic options that could produce durable remissions with a reasonable safety profile. The approval of chimeric antigen receptor (CAR) T-cell therapies has been revolutionary in the field because CAR T-cells meet this need for a substantial number of patients. With multiple approved CAR T-cell products and more expected soon, it can be difficult to distinguish between the various products and decide which to use. Effective CAR T-cell therapeutic choice is enhanced by an understanding of the biology of CAR T-cell, as well as the mechanisms associated with both efficacy and toxicity. Areas Covered: Biology of CAR T-cells, as well as a discussion of their efficacy and toxicity. Mechanisms of resistance, current unanswered questions in the field, issues associated with choosing a CAR T-cell product, and future directions for the advancement of CAR T-cell therapy. Expert Opinion: Due to differences in study populations and manufacturing times, it is too early to know if there is a 'best' choice for CAR T-cell therapy. Decisions must be individualized taking into account patient factors and expected toxicity.


Subject(s)
Immunotherapy, Adoptive , Lymphoma, B-Cell/therapy , Lymphoma, Non-Hodgkin/therapy , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes/transplantation , Antigens, CD19/immunology , B-Lymphocytes/immunology , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology
15.
Hematol Oncol Clin North Am ; 33(4): 687-705, 2019 08.
Article in English | MEDLINE | ID: mdl-31229163

ABSTRACT

Despite the myriad of available treatments, a substantial subset of patients with non-Hodgkin lymphoma are not able to achieve a prolonged disease-free interval with conventional chemotherapy or targeted agents. For these patients, hematopoietic stem cell transplantation remains an option for consolidative or curative treatment. Additionally, chimeric antigen receptor T-cell therapy has emerged for patients with relapsed/refractory B-cell lymphomas. Published studies vary widely in their selected approach to transplant and cellular therapies. This review summarizes available data related to allogeneic hematopoietic stem cell transplantation and chimeric antigen receptor T-cell therapy for the treatment of non-Hodgkin lymphomas.


Subject(s)
Immunotherapy, Adoptive/methods , Lymphoma, Non-Hodgkin/therapy , Receptors, Chimeric Antigen/therapeutic use , Stem Cell Transplantation/methods , Humans , Lymphoma, Non-Hodgkin/mortality , Survival Analysis , Transplantation, Homologous
16.
J Natl Cancer Inst ; 111(7): 646-654, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30753567

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is a revolutionary new form of immunotherapy for the treatment of hematologic malignancies. The two primary toxicities associated with CAR T-cell therapy include cytokine-release syndrome and neurotoxicity. Cytokine-release syndrome is generally self-limited but high-grade toxicities like hypotension and hypoxemia can be managed with agents that block the effects of IL-6, like tocilizumab, and/or corticosteroids. Although CAR T-cell therapy-associated neurotoxicity is a well-described clinical phenomenon, its pathophysiology remains inadequately understood; treatments and preventive strategies remain elusive. Animal models and clinical trial experience suggest the centrality of monocytes, endothelial dysfunction, and the blood-brain barrier in the development of CAR T-cell-associated neurotoxicity. Here we report what is known from preclinical models, clinical trials, and histopathologic studies regarding the pathophysiology of neurotoxicity, predictors of its incidence, and potential targets for the treatment and prevention of neurotoxicity.


Subject(s)
Hematologic Neoplasms/drug therapy , Hypotension/drug therapy , Hypoxia/drug therapy , Neurotoxicity Syndromes/drug therapy , Adrenal Cortex Hormones/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Hematologic Neoplasms/complications , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Hypotension/chemically induced , Hypoxia/chemically induced , Immunotherapy, Adoptive/adverse effects , Interleukin-6/antagonists & inhibitors , Monocytes/drug effects , Monocytes/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use
17.
J Thromb Thrombolysis ; 44(3): 392-398, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28836150

ABSTRACT

The risk benefit decision in providing anticoagulation for patients with brain metastases is amongst the most difficult decisions faced by clinicians. The purpose of our study was to evaluate both the risk of intracerebral hemorrhage (ICH) associated with anticoagulation therapy and the effect of anticoagulation on survival in patients with brain metastases and venous thromboembolism (VTE). A systematic review of the literature was performed via the PubMed, EMBASE, and the Cochrane databases. Our initial search resulted in 1304 unique citations, and 5 studies satisfied all eligibility criteria and were included for analysis. The odds ratio for development of ICH in the setting of anticoagulation was 1.37 (CI 0.86-2.17, p = 0.18). The hazard ratio for survival was 0.96 (CI 0.51-1.81, p = 0.90). While limited, the best available evidence suggests that there is no increased risk of ICH and no survival benefit associated with providing anticoagulation to patients with brain metastases who develop VTE. These patients merit individualized discussion of the risk and benefit of anticoagulation therapy. Current guidelines should be updated to include more recent studies and highlight the uncertainty of the net clinical benefit associated with anticoagulation.


Subject(s)
Anticoagulants/therapeutic use , Brain Neoplasms/secondary , Venous Thromboembolism/drug therapy , Anticoagulants/adverse effects , Brain Neoplasms/mortality , Cerebral Hemorrhage/chemically induced , Humans , Risk Assessment , Survival Analysis , Venous Thromboembolism/mortality
18.
Clin Lymphoma Myeloma Leuk ; 17(3): 145-151, 2017 03.
Article in English | MEDLINE | ID: mdl-27998707

ABSTRACT

BACKGROUND: The standard of care for diffuse large B-cell lymphoma (DLBCL) relapsing after front-line therapy is high-dose chemotherapy and autologous stem cell transplantation (ASCT). Evidence has suggested that early relapses (ie, within 1 year) after this approach portends exceptionally poor outcomes. However, data examining relapses > 1 year after ASCT for patients with refractory or relapsed DLBCL are limited, in particular, in the rituximab era. We sought to examine the effect of early (≤ 1 year) and late (> 1 year) relapse after ASCT in a single-institution cohort of patients with relapsed and refractory DLBCL treated with chemoimmunotherapy. MATERIALS AND METHODS: A retrospective analysis was performed on the data from 85 consecutive patients who had undergone ASCT for biopsy-confirmed relapsed or refractory DLBCL from 2001 to 2010 at the University of Rochester Medical Center. All patients had received rituximab as a part of treatment. Of the 85 patients, 35 developed relapse after ASCT. These 35 patients were divided into 2 groups according to the timing of the relapse (≤ 1 year and > 1 year after ASCT). RESULTS: The median follow-up period was 6.4 years. For all patients, the overall survival (OS) from post-ASCT relapse was 5.2 years. For the 27 patients developing relapse at ≤ 1 year after ASCT, the median OS was 0.6 year and progression-free survival was 0.4 year. For the 8 patients developing relapse at > 1 year after ASCT, the median OS was 5.9 years and progression-free survival was 2.9 years. CONCLUSION: Patients with relapsed or refractory DLBCL experiencing relapse > 1 year after ASCT had good outcomes. Despite the relative rarity in incidence, a significant risk of relapse of DLBCL after ASCT remains, suggesting the need for continued monitoring because of the possibility of later progression.


Subject(s)
Antineoplastic Agents/administration & dosage , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/surgery , Neoplasm Recurrence, Local/pathology , Rituximab/administration & dosage , Adult , Aged , Combined Modality Therapy/methods , Disease Progression , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Middle Aged , Retrospective Studies , Stem Cell Transplantation/methods , Transplantation, Autologous/methods , Young Adult
19.
Leuk Lymphoma ; 55(8): 1822-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24237578

ABSTRACT

Pleural effusions are common in Hodgkin lymphoma (HL). However, little is known about their prognostic significance. One hundred and ten patients with HL who presented to the University of Rochester from 1 January 2003 to 12 December 2010 were reviewed. Pleural effusions were evaluated on review of diagnostic-quality computed tomography (CT) scans. Pleural effusions were present in 26/110 patients: 1/7 (14%) stage I, 11/61 (18%) stage II, 3/18 (17%) stage III and 11/24 (46%) stage IV, and 25/91 (27%) patients had mediastinal involvement, 16/38 (42%) patients had extranodal involvement (any) and 5/14 (35%) patients had E lesions (direct extension to extranodal tissue). Unilateral and bilateral pleural effusions were equally prevalent. Survival analysis demonstrated decreased overall survival for patients with pleural effusions of borderline significance for stage I-IV (p = 0.055) but failed to show significance for patients with stage I-III (p = 0.115). Increasing stage, any extranodal involvement and bulky mediastinal disease were each predictive of pleural effusions. The presence of pleural effusion at presentation may be predictive of inferior survival for patients with Hodgkin lymphoma.


Subject(s)
Hodgkin Disease/complications , Pleural Effusion, Malignant/etiology , Female , Hodgkin Disease/diagnosis , Hodgkin Disease/mortality , Humans , Incidence , Male , Neoplasm Staging , Odds Ratio , Pleural Effusion, Malignant/diagnosis , Pleural Effusion, Malignant/epidemiology , Positron-Emission Tomography , Prognosis , Tomography, X-Ray Computed
20.
J Relig Health ; 52(3): 851-63, 2013 Sep.
Article in English | MEDLINE | ID: mdl-21882057

ABSTRACT

This study utilizes a combination of intrinsic and extrinsic Religious Orientation Scales to explore the connection between religion and health in a sample of physically active, older adults. The revised Religious Orientation Scale and the RAND Short Form 36 (SF-36) were adopted to relate religious orientation (intrinsic, extrinsic, pro-religious, and non-religious) and self-rated mental and physical health status. Individuals of pro-religious orientation reported significantly worse health for physical functioning, role limitations due to physical health, and energy or fatigue when compared with those of all other religious orientations; however, no dose-response relationships were found between religious orientation and self-rated health. The results of this study indicate that deleterious health effects may accompany pro-religious orientation. Caution is provided for directors of religious programs for older adults.


Subject(s)
Health Status , Religion and Psychology , Religion , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Regression Analysis , Surveys and Questionnaires , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...