Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Dev Sci ; 25(4): e13219, 2022 07.
Article in English | MEDLINE | ID: mdl-34935245

ABSTRACT

Studies on children's understanding of counting examine when and how children acquire the cardinal principle: the idea that the last word in a counted set reflects the cardinal value of the set. Using Wynn's (1990) Give-N Task, researchers classify children who can count to generate large sets as having acquired the cardinal principle (cardinal-principle-knowers) and those who cannot as lacking knowledge of it (subset-knowers). However, recent studies have provided a more nuanced view of number word acquisition. Here, we explore this view by examining the developmental progression of the counting principles with an aim to elucidate the gradual elements that lead to children successfully generating sets and being classified as CP-knowers on the Give-N Task. Specifically, we test the claim that subset-knowers lack cardinal principle knowledge by separating children's understanding of the cardinal principle from their ability to apply and implement counting procedures. We also ask when knowledge of Gelman & Gallistel's (1978) other how-to-count principles emerge in development. We analyzed how often children violated the three how-to-count principles in a secondary analysis of Give-N data (N = 86). We found that children already have knowledge of the cardinal principle prior to becoming CP-knowers, and that understanding of the stable-order and word-object correspondence principles likely emerged earlier. These results suggest that gradual development may best characterize children's acquisition of the counting principles and that learning to coordinate all three principles represents an additional step beyond learning them individually.


Subject(s)
Concept Formation , Problem Solving , Child , Child Development , Humans , Knowledge , Learning
2.
Exerc Immunol Rev ; 26: 56-78, 2020.
Article in English | MEDLINE | ID: mdl-32139349

ABSTRACT

PURPOSE: Habitual intense exercise may increase the incidence of upper respiratory symptoms (URS) in elite athletes. This study investigated whether immune gene expression could identify gene markers that discriminate athletes with a higher prevalence of URS. METHODS: This cross-sectional analysis of elite Australian athletes from various sports investigated whether athletes retrospectively reporting URS for two days or more in a month (n=38), had an altered immune gene expression profile compared with asymptomatic athletes (n=33). Peripheral blood samples were collected during Olympic selection events with corresponding URS data collected for the one-month period before sampling. Digital immune gene expression analysis was undertaken using the NanoString PanCancer Immune Profiling panel. RESULTS: Fifty immune genes were differentially expressed between the groups (p<0.05) and approximately 78% of these genes were more highly expressed in athletes reporting URS. Many of these genes were interferon-stimulated genes or genes involved in the Jak/Stat signalling pathway. Only interferon alpha inducible protein 27 (IFI27), an interferon stimulated gene involved in viral response, remained significantly higher in athletes reporting URS (log2 fold-difference=2.49, odds ratio 1.02 per unit increase; p<0.01) post-adjustment and discriminated athletes reporting URS from asymptomatic athletes with 78% accuracy. CONCLUSIONS: Expression of IFI27 could differentiate athletes reporting URS from asymptomatic athletes, a gene that is upregulated in the immune response to viral infection. Upregulation of viral signalling pathways provides novel information on the potential aetiology of URS in elite Olympic athletes.


Subject(s)
Athletes , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/genetics , Transcriptome , Australia , Cross-Sectional Studies , Humans , Membrane Proteins/genetics , Retrospective Studies
3.
Br J Sports Med ; 52(1): 47-53, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29056598

ABSTRACT

OBJECTIVE: Establish the prevalence of illness symptoms, poor sleep quality, poor mental health symptoms, low energy availability and stress-recovery state in an Olympic cohort late in the 3 months prior to the Summer Olympic Games. METHODS: Olympic athletes (n=317) from 11 sports were invited to complete questionnaires administered 3 months before the Rio 2016 Olympic Games. These questionnaires included the Depression, Anxiety and Stress Questionnaire, Perceived Stress Scale, Dispositional Resilience Scale, Recovery-Stress Questionnaire (REST-Q-52 item), Low Energy Availability in Females Questionnaire (LEAF-Q), Epworth Sleepiness Scale, Pittsburgh Sleep Quality Index and custom-made questionnaires on probiotic usage and travel. Multiple illness (case) definitions were applied. ORs and attributable fractions in the population were used. Factor analyses were used to explore the relationships between variables. RESULTS: The response rate was of 42% (male, n=47, age 25.8±4.1 years; female, n=85, age 24.3±3.9 years). Low energy availability was associated with sustaining an illness in the previous month (upper respiratory, OR=3.8, 95% CI 1.2 to 12). The main factor relating to illness pertained to a combination of anxiety and stress-recovery states (as measured by the REST-Q-52 item). All participants reported at least one episode of illness in the last month (100% prevalence). CONCLUSIONS: All participants reported at least one illness symptom in the previous month. Low energy availability was a leading variable associated with illness in Olympic-class athletes. The estimates duration of symptoms ranged from 2 to 7 days. Factor analyses show the interdependence of various health domains and support multidisciplinary care.


Subject(s)
Athletes/psychology , Fatigue/epidemiology , Mental Health , Sleep , Adult , Anniversaries and Special Events , Anxiety , Cross-Sectional Studies , Female , Health Status , Humans , Male , Prevalence , Self Report , Stress, Psychological , Surveys and Questionnaires , Young Adult
4.
J Clin Oncol ; 35(29): 3322-3329, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-28809608

ABSTRACT

Purpose Adoptive transfer of genetically modified T cells is being explored as a treatment for patients with metastatic cancer. Most current strategies use genes that encode major histocompatibility complex (MHC) class I-restricted T-cell receptors (TCRs) or chimeric antigen receptors to genetically modify CD8+ T cells or bulk T cells for treatment. Here, we evaluated the safety and efficacy of an adoptive CD4+ T-cell therapy using an MHC class II-restricted, HLA-DPB1*0401-restricted TCR that recognized the cancer germline antigen, MAGE-A3 (melanoma-associated antigen-A3). Patients and Methods Patients received a lymphodepleting preparative regimen, followed by adoptive transfer of purified CD4+ T cells, retrovirally transduced with MAGE-A3 TCR plus systemic high-dose IL-2. A cell dose escalation was conducted, starting at 107 total cells and escalating at half-log increments to approximately 1011 cells. Nine patients were treated at the highest dose level (0.78 to 1.23 × 1011 cells). Results Seventeen patients were treated. During the cell dose-escalation phase, an objective complete response was observed in a patient with metastatic cervical cancer who received 2.7 × 109 cells (ongoing at ≥ 29 months). Among nine patients who were treated at the highest dose level, objective partial responses were observed in a patient with esophageal cancer (duration, 4 months), a patient with urothelial cancer (ongoing at ≥ 19 months), and a patient with osteosarcoma (duration, 4 months). Most patients experienced transient fevers and the expected hematologic toxicities from lymphodepletion pretreatment. Two patients experienced transient grade 3 and 4 transaminase elevations. There were no treatment-related deaths. Conclusion These results demonstrate the safety and efficacy of administering autologous CD4+ T cells that are genetically engineered to express an MHC class II-restricted antitumor TCR that targets MAGE-A3. This clinical trial extends the reach of TCR gene therapy for patients with metastatic cancer.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/transplantation , Genetic Therapy/methods , HLA-DP beta-Chains/immunology , Immunotherapy, Adoptive/methods , Neoplasm Proteins/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Administration, Intravenous , Aged , Antineoplastic Agents/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Female , Genetic Therapy/adverse effects , Humans , Immunotherapy, Adoptive/adverse effects , Interleukin-2/administration & dosage , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/genetics , Time Factors , Transplantation, Autologous , Treatment Outcome
5.
J Sci Med Sport ; 20(8): 745-750, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28385561

ABSTRACT

OBJECTIVES: Illness can disrupt training and competition performance of athletes. Few studies have quantified the relative contribution of the known medical, behavioural and lifestyle risk factors. DESIGN: Cross-sectional. METHODS: Olympic athletes from 11 sports (n=221) were invited to complete questionnaires administered nine months before the Rio 2016 Olympic Games. These included the Depression, Anxiety and Stress Questionnaire (DASS-21), Perceived Stress Scale (PSS), Dispositional Resilience Scale (DRS), Recovery-Stress Questionnaire (REST-Q-52 item), Low Energy in Females Questionnaire (LEAF-Q), a modified Personal and Household Hygiene questionnaire, Epworth Sleepiness Scale, Pittsburgh Sleep Quality Index, and custom-made questionnaires on probiotic usage and travel. An illness (case) was defined as an event which limited training or competition for greater hours in the prior month. Odds ratios and attributable fractions in the population (AFP) were utilised for categorical variables with independent t-tests or Wilcoxon rank-sum for continuous variables. RESULTS: Eighty-one athletes responded (male, n=26; female, n=55). There were 16 illness cases and 65 controls. Female athletes were at higher odds of illness (OR=9.4, 95%CI 1.3-410, p=0.01, AFP=0.84). Low energy availability (LEAF-Q score ≥8: OR=7.4, 95%CI 0.78-352, p=0.04, AFP=0.76), depression symptoms (DASS-21: depression score >4, OR=8.4, 95%CI 1.1-59, p<0.01; AFP=0.39) and higher perceived stress (PSS: 10-item, p=0.04) were significantly associated with illness. CONCLUSIONS: Female sex, low energy availability, and mental health are associated with sports incapacity (time loss) due to illness. Low energy availability had high attributable fractions in the population and stands out as a primary association with illness.


Subject(s)
Athletes/psychology , Health Status Indicators , Sports Medicine/statistics & numerical data , Adult , Case-Control Studies , Cross-Sectional Studies , Depression/epidemiology , Female , Humans , Male , Neuropsychological Tests , Risk Factors , Sex Factors , Stress, Psychological/epidemiology , Surveys and Questionnaires , Young Adult
6.
Lancet Oncol ; 18(6): 792-802, 2017 06.
Article in English | MEDLINE | ID: mdl-28395880

ABSTRACT

BACKGROUND: Uveal melanoma is a rare tumour with no established treatments once metastases develop. Although a variety of immune-based therapies have shown efficacy in metastatic cutaneous melanoma, their use in ocular variants has been disappointing. Recently, adoptive T-cell therapy has shown salvage responses in multiple refractory solid tumours. Thus, we sought to determine if adoptive transfer of autologous tumour-infiltrating lymphocytes (TILs) could mediate regression of metastatic uveal melanoma. METHODS: In this ongoing single-centre, two-stage, phase 2, single-arm trial, patients (aged ≥16 years) with histologically confirmed metastatic ocular melanoma were enrolled. Key eligibility criteria were an Eastern Cooperative Oncology Group performance status of 0 or 1, progressive metastatic disease, and adequate haematological, renal, and hepatic function. Metastasectomies were done to procure tumour tissue to generate autologous TIL cultures, which then underwent large scale ex-vivo expansion. Patients were treated with lymphodepleting conditioning chemotherapy (intravenous cyclophosphamide [60 mg/kg] daily for 2 days followed by fludarabine [25 mg/m2] daily for 5 days, followed by a single intravenous infusion of autologous TILs and high-dose interleukin-2 [720 000 IU/kg] every 8 h). The primary endpoint was objective tumour response in evaluable patients per protocol using Response to Evaluation Criteria in Solid Tumors, version 1.0. An interim analysis of this trial is reported here. The trial is registered at ClinicalTrials.gov, number NCT01814046. FINDINGS: From the completed first stage and ongoing expansion stage of this trial, a total of 21 consecutive patients with metastatic uveal melanoma were enrolled between June 7, 2013, and Sept 9, 2016, and received TIL therapy. Seven (35%, 95% CI 16-59) of 20 evaluable patients had objective tumour regression. Among the responders, six patients achieved a partial response, two of which are ongoing and have not reached maximum response. One patient achieved complete response of numerous hepatic metastases, currently ongoing at 21 months post therapy. Three of the responders were refractory to previous immune checkpoint blockade. Common grade 3 or worse toxic effects were related to the lymphodepleting chemotherapy regimen and included lymphopenia, neutropenia, and thrombocytopenia (21 [100%] patients for each toxicity); anaemia (14 [67%] patients); and infection (six [29%] patients). There was one treatment-related death secondary to sepsis-induced multiorgan failure. INTERPRETATION: To our knowledge, this is the first report describing adoptive transfer of autologous TILs to mediate objective tumour regression in patients with metastatic uveal melanoma. These initial results challenge the belief that metastatic uveal melanoma is immunotherapy resistant and support the further investigation of immune-based therapies for this cancer. Refinement of this T-cell therapy is crucial to improve the frequency of clinical responses and the general applicability of this treatment modality. FUNDING: Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research.


Subject(s)
Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma/therapy , Uveal Neoplasms/therapy , Adult , Anemia/chemically induced , Eye Enucleation , Female , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Infections/chemically induced , Lymphopenia/chemically induced , Male , Melanoma/genetics , Melanoma/secondary , Metastasectomy , Middle Aged , Neutropenia/chemically induced , Radiotherapy , Response Evaluation Criteria in Solid Tumors , Thrombocytopenia/chemically induced , Transplantation Conditioning/adverse effects , Transplantation, Autologous , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
7.
J Clin Oncol ; 35(16): 1803-1813, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28291388

ABSTRACT

Purpose T cells genetically modified to express chimeric antigen receptors (CARs) targeting CD19 (CAR-19) have potent activity against acute lymphoblastic leukemia, but fewer results supporting treatment of lymphoma with CAR-19 T cells have been published. Patients with lymphoma that is chemotherapy refractory or relapsed after autologous stem-cell transplantation have a grim prognosis, and new treatments for these patients are clearly needed. Chemotherapy administered before adoptive T-cell transfer has been shown to enhance the antimalignancy activity of adoptively transferred T cells. Patients and Methods We treated 22 patients with advanced-stage lymphoma in a clinical trial of CAR-19 T cells preceded by low-dose chemotherapy. Nineteen patients had diffuse large B-cell lymphoma, two patients had follicular lymphoma, and one patient had mantle cell lymphoma. Patients received a single dose of CAR-19 T cells 2 days after a low-dose chemotherapy conditioning regimen of cyclophosphamide plus fludarabine. Results The overall remission rate was 73% with 55% complete remissions and 18% partial remissions. Eleven of 12 complete remissions are ongoing. Fifty-five percent of patients had grade 3 or 4 neurologic toxicities that completely resolved. The low-dose chemotherapy conditioning regimen depleted blood lymphocytes and increased serum interleukin-15 (IL-15). Patients who achieved a remission had a median peak blood CAR+ cell level of 98/µL and those who did not achieve a remission had a median peak blood CAR+ cell level of 15/µL ( P = .027). High serum IL-15 levels were associated with high peak blood CAR+ cell levels ( P = .001) and remissions of lymphoma ( P < .001). Conclusion CAR-19 T cells preceded by low-dose chemotherapy induced remission of advanced-stage lymphoma, and high serum IL-15 levels were associated with the effectiveness of this treatment regimen. CAR-19 T cells will likely become an important treatment for patients with relapsed lymphoma.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-15/blood , Lymphoma/therapy , T-Lymphocytes/transplantation , Adult , Aged , Antigens, CD19/immunology , Cyclophosphamide/administration & dosage , Humans , Interleukin-15/immunology , Lymphoma/blood , Lymphoma/drug therapy , Lymphoma/immunology , Middle Aged , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transplantation Conditioning/methods , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
8.
J Clin Oncol ; 34(20): 2389-97, 2016 07 10.
Article in English | MEDLINE | ID: mdl-27217459

ABSTRACT

PURPOSE: Adoptive cell transfer, the infusion of large numbers of activated autologous lymphocytes, can mediate objective tumor regression in a majority of patients with metastatic melanoma (52 of 93; 56%). Addition and intensification of total body irradiation (TBI) to the preparative lymphodepleting chemotherapy regimen in sequential trials improved objective partial and complete response (CR) rates. Here, we evaluated the importance of adding TBI to the adoptive transfer of tumor-infiltrating lymphocytes (TIL) in a randomized fashion. PATIENTS AND METHODS: A total of 101 patients with metastatic melanoma, including 76 patients with M1c disease, were randomly assigned to receive nonmyeloablative chemotherapy with or without 1,200 cGy TBI before transfer of tumor-infiltrating lymphcytes. Primary end points were CR rate (as defined by Response Evaluation Criteria in Solid Tumors v1.0) and overall survival (OS). Clinical and laboratory data were analyzed for correlates of response. RESULTS: CR rates were 24% in both groups (12 of 50 v 12 of 51), and OS was also similar (median OS, 38.2 v 36.6 months; hazard ratio, 1.11; 95% CI, 0.65 to 1.91; P = .71). Thrombotic microangiopathy was an adverse event unique to the TBI arm and occurred in 13 of 48 treated patients. With a median potential follow-up of 40.9 months, only one of 24 patients who achieved a CR recurred. CONCLUSION: Adoptive cell transfer can mediate durable complete regressions in 24% of patients with metastatic melanoma, with median survival > 3 years. Results were similar using chemotherapy preparative regimens with or without addition of TBI.


Subject(s)
Immunotherapy, Adoptive , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , Adolescent , Adult , Aged , Female , Humans , Male , Melanoma/immunology , Melanoma/mortality , Middle Aged , Neoplasm Metastasis , Prospective Studies , Whole-Body Irradiation
9.
Ann Surg Oncol ; 23(4): 1309-19, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26597368

ABSTRACT

PURPOSE: There is no consensus for the treatment of melanoma metastatic to the liver. Percutaneous hepatic perfusion with melphalan (PHP-Mel) is a method of delivering regional chemotherapy selectively to the liver. In this study, we report the results of a multicenter, randomized controlled trial comparing PHP-Mel with best alternative care (BAC) for patients with ocular or cutaneous melanoma metastatic to the liver. PATIENTS AND METHODS: A total of 93 patients were randomized to PHP-Mel (n = 44) or BAC (n = 49). On the PHP-Mel arm, melphalan was delivered via the hepatic artery, and the hepatic effluent captured and filtered extracorporeally prior to return to the systemic circulation via a venovenous bypass circuit. PHP-Mel was repeatable every 4-8 weeks. The primary endpoint was hepatic progression-free survival (hPFS), and secondary endpoints included overall PFS (oPFS), overall survival (OS), hepatic objective response (hOR), and safety. RESULTS: hPFS was 7.0 months for PHP-Mel and 1.6 months for BAC (p < 0.0001), while oPFS was 5.4 months for PHP-Mel and 1.6 months for BAC (p < 0.0001). Median OS was not significantly different (PHP-Mel 10.6 months vs. BAC 10.0 months), likely due to crossover to PHP-Mel treatment (57.1 %) from the BAC arm, and the hOR was 36.4 % for PHP-Mel and 2.0 % for BAC (p < 0.001). The majority of adverse events were related to bone marrow suppression. Four deaths were attributed to PHP-Mel, three in the primary PHP-Mel group, and one post-crossover to PHP-Mel from BAC. CONCLUSION: This randomized, phase III study demonstrated the efficacy of the PHP-Mel procedure. hPFS, oPFS, and hOR were significantly improved with PHP-Mel. PHP with melphalan should provide a new treatment option for unresectable metastatic melanoma in the liver.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Eye Neoplasms/secondary , Hepatic Artery , Liver Neoplasms/secondary , Melanoma/pathology , Skin Neoplasms/secondary , Adult , Aged , Chemotherapy, Cancer, Regional Perfusion , Embolization, Therapeutic , Eye Neoplasms/drug therapy , Female , Follow-Up Studies , Humans , Infusions, Intra-Arterial , Liver Neoplasms/drug therapy , Male , Melanoma/drug therapy , Middle Aged , Neoplasm Staging , Perfusion , Prognosis , Skin Neoplasms/drug therapy , Survival Rate
10.
Clin Cancer Res ; 21(10): 2278-88, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25695689

ABSTRACT

PURPOSE: Infusion of interleukin-12 (IL12) can mediate antitumor immunity in animal models, yet its systemic administration to patients with cancer results in minimal efficacy and severe toxicity. Here, we evaluated the antitumor activity of adoptively transferred human tumor-infiltrating lymphocytes (TILs) genetically engineered to secrete single-chain IL12 selectively at the tumor site. EXPERIMENTAL DESIGN: Thirty-three patients with metastatic melanoma were treated in a cell dose-escalation trial of autologous TILs transduced with a gene encoding a single-chain IL12 driven by a nuclear factor of the activated T cells promoter (NFAT.IL12). No IL2 was administered. RESULTS: The administration of 0.001 to 0.1 × 10(9) NFAT.IL12-transduced TILs to 17 patients resulted in a single, objective response (5.9%). However, at doses between 0.3 and 3 × 10(9) cells, 10 of 16 patients (63%) exhibited objective clinical responses. The responses tended to be short, and the administered IL12-producing cells rarely persisted at 1 month. Increasing cell doses were associated with high serum levels of IL12 and IFNγ as well as clinical toxicities, including liver dysfunction, high fevers, and sporadic life-threatening hemodynamic instability. CONCLUSIONS: In this first-in-man trial, administration of TILs transduced with an inducible IL12 gene mediated tumor responses in the absence of IL2 administration using cell doses 10- to 100-fold lower than conventional TILs. However, due to toxicities, likely attributable to the secreted IL12, further refinement will be necessary before this approach can be safely used in the treatment of cancer patients.


Subject(s)
Interleukin-12/genetics , Lymphocytes, Tumor-Infiltrating/physiology , Melanoma/therapy , Skin Neoplasms/therapy , Adult , Aged , Cells, Cultured , Female , Genetic Engineering , Humans , Immunotherapy , Interleukin-12/biosynthesis , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Melanoma/immunology , Melanoma/secondary , Middle Aged , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Transcriptional Activation , Treatment Outcome , Young Adult
11.
J Clin Oncol ; 33(6): 540-9, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25154820

ABSTRACT

PURPOSE: T cells can be genetically modified to express an anti-CD19 chimeric antigen receptor (CAR). We assessed the safety and efficacy of administering autologous anti-CD19 CAR T cells to patients with advanced CD19(+) B-cell malignancies. PATIENTS AND METHODS: We treated 15 patients with advanced B-cell malignancies. Nine patients had diffuse large B-cell lymphoma (DLBCL), two had indolent lymphomas, and four had chronic lymphocytic leukemia. Patients received a conditioning chemotherapy regimen of cyclophosphamide and fludarabine followed by a single infusion of anti-CD19 CAR T cells. RESULTS: Of 15 patients, eight achieved complete remissions (CRs), four achieved partial remissions, one had stable lymphoma, and two were not evaluable for response. CRs were obtained by four of seven evaluable patients with chemotherapy-refractory DLBCL; three of these four CRs are ongoing, with durations ranging from 9 to 22 months. Acute toxicities including fever, hypotension, delirium, and other neurologic toxicities occurred in some patients after infusion of anti-CD19 CAR T cells; these toxicities resolved within 3 weeks after cell infusion. One patient died suddenly as a result of an unknown cause 16 days after cell infusion. CAR T cells were detected in the blood of patients at peak levels, ranging from nine to 777 CAR-positive T cells/µL. CONCLUSION: This is the first report to our knowledge of successful treatment of DLBCL with anti-CD19 CAR T cells. These results demonstrate the feasibility and effectiveness of treating chemotherapy-refractory B-cell malignancies with anti-CD19 CAR T cells. The numerous remissions obtained provide strong support for further development of this approach.


Subject(s)
Antigens, CD19/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Adult , Aged , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Female , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/immunology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Male , Middle Aged , T-Lymphocytes/immunology , Transplantation Conditioning/methods , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
12.
J Surg Oncol ; 110(3): 275-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25042700

ABSTRACT

BACKGROUND: A prospective randomized trial was conducted to compare the impact of systemic chemotherapy versus multi-modality therapy (complete cytoreductive surgery (CRS), hyperthermic intraperitoneal chemotherapy (HIPEC), and systemic chemotherapy) on overall survival (OS) in patients with gastric carcinomatosis. METHODS: Patients with measurable metastatic gastric adenocarcinoma involving the peritoneum, and resectable to "no evidence of disease" were randomized to gastrectomy, metastasectomy, HIPEC, and systemic FOLFOXIRI (GYMS arm) or FOLFOXIRI alone (SA arm). RESULTS: Seventeen patients were enrolled (16 evaluable); 7 of 9 patients in the multi-modality GYMS arm achieved complete cytoreduction (CCR0). Median OS was 11.3 months in the GYMS arm and 4.3 months in the SA arm. Four patients in the GYMS arm survived >12 months, 2 patients close to 2 years at last follow-up, and 1 patient more than 4 years, with 2 of these patients still alive. No patient in the SA arm lived beyond 11 months. All patients surviving beyond 12 months in the surgery arm achieved complete cytoreduction and had an initial Peritoneal Cancer Index (PCI) of ≤ 15. CONCLUSION: Maximal cytoreductive surgery combined with regional (HIPEC) and systemic chemotherapy in selected patients with gastric carcinomatosis and limited disease burden can achieve prolonged survival.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/therapy , Chemotherapy, Cancer, Regional Perfusion , Hyperthermia, Induced , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/therapy , Stomach Neoplasms/mortality , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Female , Fluorouracil/therapeutic use , Gastrectomy , Humans , Kaplan-Meier Estimate , Leucovorin/therapeutic use , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Middle Aged , Organoplatinum Compounds/therapeutic use , Peritoneal Neoplasms/secondary , Peritoneum/surgery , Prospective Studies , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy
13.
Trials ; 12: 129, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21595953

ABSTRACT

BACKGROUND: Pancreatic cancer is the fourth leading cause of cancer death in the United States. Surgery offers the only chance for cure. However, less than twenty percent of patients are considered operative candidates at the time of diagnosis. A common reason for being classified as unresectable is advanced loco-regional disease.A review of the literature indicates that almost nine hundred patients with pancreatic cancer have received regional chemotherapy in the last 15 years. Phase I studies have shown regional administration of chemotherapy to be safe. The average reported response rate was approximately 26%. The average 1-year survival was 39%, with an average median survival of 9 months. Of the patients that experienced a radiographic response to therapy, 78 (78/277, 28%) patients underwent exploratory surgery following regional chemotherapy administration; thirty-two (41%) of those patients were amenable to pancreatectomy. None of the studies performed analyses to identify factors predicting response to regional chemotherapy.Progressive surgical techniques combined with current neoadjuvant chemoradiotherapy strategies have already yielded emerging support for a multimodality approach to treatment of advanced pancreatic cancer.Intravenous gemcitabine is the current standard treatment of pancreatic cancer. However, >90% of the drug is secreted unchanged affecting toxicity but not the cancer per se. Gemcitabine is converted inside the cell into its active drug form in a rate limiting reaction. We hypothesize that neoadjuvant regional chemotherapy with continuous infusion of gemcitabine will be well tolerated and may improve resectability rates in cases of locally advanced pancreatic cancer. DESIGN: This is a phase I study designed to evaluate the feasibility and toxicity of super-selective intra-arterial administration of gemcitabine in patients with locally advanced, unresectable pancreatic adenocarcinoma. Patients considered unresectable due to locally advanced pancreatic cancer will receive super-selective arterial infusion of gemcitabine over 24 hours via subcutaneous indwelling port. Three to six patients will be enrolled per dose cohort, with seven cohorts, plus an additional six patients at the maximum tolerated dose; accrual is expected to last 36 months. Secondary objectives will include the determination of progression free and overall survival, as well as the conversion rate from unresectable to potentially resectable pancreatic cancer. TRIAL REGISTRATION: ClinicalTrials.gov ID: NCT01294358.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/administration & dosage , Chemotherapy, Cancer, Regional Perfusion , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Research Design , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Antimetabolites, Antineoplastic/adverse effects , Chemotherapy, Adjuvant , Chemotherapy, Cancer, Regional Perfusion/adverse effects , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Embolization, Therapeutic , Humans , Maryland , Neoadjuvant Therapy , Neoplasm Invasiveness , Pancreatectomy , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Treatment Outcome , Gemcitabine
14.
Trials ; 11: 62, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20500867

ABSTRACT

BACKGROUND: The standard of care for colorectal peritoneal carcinomatosis is evolving from chemotherapy to cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) for patients with disease limited to the peritoneum. Peritoneal carcinomatosis from colorectal cancer treated with chemotherapy alone results in median survival of 5 to 13 months, whereas CRS with HIPEC for early peritoneal carcinomatosis from colorectal cancer resulted in median survival of 48-63 months and 5 year survival of 51%.Completeness of cytoreduction and limited disease are associated with longer survival, yet early peritoneal carcinomatosis is undetectable by conventional imaging. Exploratory laparotomy can successfully identify early disease, but this approach can only be justified in patients with high risk of peritoneal carcinomatosis. Historical data indicates that patients presenting with synchronous peritoneal carcinomatosis, ovarian metastases, perforated primary tumor, and emergency presentation with bleeding or obstructing lesions are at high risk of peritoneal carcinomatosis. Approximately 55% of these patient populations will develop peritoneal carcinomatosis. We hypothesize that performing a mandatory second look laparotomy with CRS and HIPEC for patients who are at high risk for developing peritoneal carcinomatosis from colorectal cancer will lead to improved survival as compared to patients who receive standard of care with routine surveillance. METHODS/DESIGN: This study is a prospective randomized trial designed to answer the question whether mandatory second look surgery with CRS and HIPEC will prolong overall survival compared to the standard of care in patients who are at high risk for developing peritoneal carcinomatosis from colorectal cancer (CRC). Patients with CRC at high risk for developing peritoneal carcinomatosis who underwent curative surgery and subsequently received standard of care adjuvant chemotherapy will be evaluated. The patients who remain without evidence of disease by imaging, physical examination, and tumor markers for 12 months after the primary operation will be randomized to mandatory second look surgery or standard-of-care surveillance. At laparotomy, CRS and HIPEC will be performed with intraperitoneal oxaliplatin with concurrent systemic 5-fluorouracil and leucovorin. Up to 100 patients will be enrolled to allow for 35 evaluable patients in each arm; accrual is expected to last 5 years. TRIAL REGISTRATION: ClinicalTrials.gov ID: NCT01095523.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Cancer, Regional Perfusion , Colorectal Neoplasms/therapy , Hyperthermia, Induced , Laparotomy , Peritoneal Neoplasms/therapy , Second-Look Surgery , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Adjuvant , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Fluorouracil/administration & dosage , Humans , Hyperthermia, Induced/adverse effects , Laparotomy/adverse effects , Leucovorin/administration & dosage , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/secondary , Prospective Studies , Risk Assessment , Risk Factors , Sample Size , Second-Look Surgery/adverse effects , Time Factors , Treatment Outcome
15.
Trials ; 10: 121, 2009 Dec 23.
Article in English | MEDLINE | ID: mdl-20030854

ABSTRACT

BACKGROUND: The standard of care for metastatic gastric cancer (MGC) is systemic chemotherapy which leads to a median survival of 6-15 months. Survival beyond 3 years is rare. For selected groups of patients with limited MGC, retrospective studies have shown improved overall survival following gastrectomy and metastasectomies including peritoneal stripping with continuous hyperthermic peritoneal perfusion (CHPP), liver resection, and pulmonary resection. Median survival after liver resection for MGC is up to 34 months, with a five year survival rate of 24.5%. Similarly, reported median survival after pulmonary resection of MGC is 21 months with long term survival of greater than 5 years a possibility. Several case reports and small studies have documented evidence of long-term survival in select individuals who undergo CHPP for MGC. DESIGN: The GYMSSA trial is a prospective randomized trial for patients with MGC. It is designed to compare two therapeutic approaches: gastrectomy with metastasectomy plus systemic chemotherapy (GYMS) versus systemic chemotherapy alone (SA). Systemic therapy will be composed of the FOLFOXIRI regimen. The aim of the study is to evaluate overall survival and potential selection criteria to determine those patients who may benefit from surgery plus systemic therapy. The study will be conducted by the Surgery Branch at the National Cancer Institute (NCI), National Institutes of Health (NIH) in Bethesda, Maryland. Surgeries and followup will be done at the NCI, and chemotherapy will be given by either the local oncologist or the medical oncology branch at NCI. TRIAL REGISTRATION: ClinicalTrials.gov ID. NCT00941655.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Gastrectomy , Hyperthermia, Induced/methods , Stomach Neoplasms , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Combined Modality Therapy , Fluorouracil/therapeutic use , Hepatectomy , Humans , Leucovorin/therapeutic use , Organoplatinum Compounds/therapeutic use , Peritoneum/surgery , Pneumonectomy , Stomach Neoplasms/drug therapy , Stomach Neoplasms/secondary , Stomach Neoplasms/surgery , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...