Network


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

Hotspot


Dive into the research topics where Kathleen Ruehle is active.

Publication


Featured researches published by Kathleen Ruehle.


Nature Medicine | 2005

Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer

Aaron P. Rapoport; Edward A. Stadtmauer; Nicole A. Aqui; Ashraf Badros; Julio Cotte; Lisa Chrisley; Elizabeth Veloso; Zhaohui Zheng; Sandra Westphal; Rebecca Mair; Nina Chi; Bashi Ratterree; Mary Francis Pochran; Sabrina Natt; Joanne Hinkle; Cheryl Sickles; Ambika Sohal; Kathleen Ruehle; Christian Lynch; Lei Zhang; David L. Porter; Selina M. Luger; Chuanfa Guo; Hong-Bin Fang; William C. Blackwelder; Kim Hankey; Dean L. Mann; Robert Edelman; Carl E. Frasch; Bruce L. Levine

Immunodeficiency is a barrier to successful vaccination in individuals with cancer and chronic infection. We performed a randomized phase 1/2 study in lymphopenic individuals after high-dose chemotherapy and autologous hematopoietic stem cell transplantation for myeloma. Combination immunotherapy consisting of a single early post-transplant infusion of in vivo vaccine-primed and ex vivo costimulated autologous T cells followed by post-transplant booster immunizations improved the severe immunodeficiency associated with high-dose chemotherapy and led to the induction of clinically relevant immunity in adults within a month after transplantation. Immune assays showed accelerated restoration of CD4 T-cell numbers and function. Early T-cell infusions also resulted in significantly improved T-cell proliferation in response to antigens that were not contained in the vaccine, as assessed by responses to staphylococcal enterotoxin B and cytomegalovirus antigens (P < 0.05). In the setting of lymphopenia, combined vaccine therapy and adoptive T-cell transfer fosters the development of enhanced memory T-cell responses.


Clinical Cancer Research | 2009

Rapid Immune Recovery and Graft-versus-Host Disease ^ like Engraftment Syndrome following Adoptive Transfer of Costimulated Autologous T Cells

Aaron P. Rapoport; Edward A. Stadtmauer; Nicole A. Aqui; Dan T. Vogl; Anne Chew; Hong-Bin Fang; Stephen Janofsky; Kelly Yager; Elizabeth Veloso; Zhaohui Zheng; Todd Milliron; Sandra Westphal; Julio Cotte; Hong Huynh; Andrea Cannon; Saul Yanovich; Gorgun Akpek; Ming Tan; Kristen Virts; Kathleen Ruehle; Carolynn Harris; Sunita Philip; Robert H. Vonderheide; Bruce L. Levine; Carl H. June

Purpose: Previously, we showed that adoptive transfer of in vivo vaccine-primed and ex vivo (anti-CD3/anti-CD28) costimulated autologous T cells (ex-T) at day +12 after transplant increased CD4 and CD8 T-cell counts at day +42 and augmented vaccine-specific immune responses in patients with myeloma. Here, we investigated the safety and kinetics of T-cell recovery after infusing ex-T at day +2 after transplant. Experimental Design: In this phase I/II two-arm clinical trial, 50 patients with myeloma received autografts after high-dose melphalan followed by infusions of ex-T at day +2 after transplant. Patients also received pretransplant and posttransplant immunizations using a pneumococcal conjugate vaccine only (arm B; n = 24) or the pneumococcal conjugate vaccine plus an HLA-A2–restricted multipeptide vaccine for HLA-A2+ patients (arm A; n = 26). Results: The mean number of T cells infused was 4.26 × 1010 (range, 1.59-5.0). At day 14 after transplant, the median CD3, CD4, and CD8 counts were 4,198, 1,545, and 2,858 cells/μL, respectively. Interleukin (IL)-6 and IL-15 levels increased early after transplant and IL-15 levels correlated significantly to day 14 T-cell counts. Robust vaccine-specific B- and T-cell responses were generated. T-cell infusions were well tolerated with no effect on hematopoietic recovery. Eight patients (16%) developed a T-cell “engraftment syndrome” characterized by diarrhea and fever that was clinically and histopathologically indistinguishable from grade 1 to 3 acute graft-versus-host disease (GVHD) of the gastrointestinal tract (seven patients) and/or grade 1 to 2 cutaneous GVHD (four patients). Conclusions: Adoptive T-cell transfers achieve robust T-cell recovery early after transplant and induce moderate-to-severe autologous GVHD in a subset of patients.


Clinical Cancer Research | 2014

Combination Immunotherapy after ASCT for Multiple Myeloma Using MAGE-A3/Poly-ICLC Immunizations Followed by Adoptive Transfer of Vaccine-Primed and Costimulated Autologous T Cells

Aaron P. Rapoport; Nicole A. Aqui; Edward A. Stadtmauer; Dan T. Vogl; Yin Yan Xu; Michael Kalos; Ling Cai; Hong-Bin Fang; Brendan M. Weiss; Ashraf Badros; Saul Yanovich; Gorgun Akpek; Patricia Tsao; Alan S. Cross; Dean L. Mann; Sunita Philip; Naseem Kerr; Andrea L. Brennan; Zhaohui Zheng; Kathleen Ruehle; Todd Milliron; Scott E. Strome; Andres M. Salazar; Bruce L. Levine; Carl H. June

Purpose: Myeloma-directed cellular immune responses after autologous stem cell transplantation (ASCT) may reduce relapse rates. We studied whether coinjecting the TLR-3 agonist and vaccine adjuvant Poly-ICLC with a MAGE-A3 peptide vaccine was safe and would elicit a high frequency of vaccine-directed immune responses when combined with vaccine-primed and costimulated autologous T cells. Experimental Design: In a phase II clinical trial (NCT01245673), we evaluated the safety and activity of ex vivo expanded autologous T cells primed in vivo using a MAGE-A3 multipeptide vaccine (compound GL-0817) combined with Poly-ICLC (Hiltonol), granulocyte macrophage colony-stimulating factor (GM-CSF) ± montanide. Twenty-seven patients with active and/or high-risk myeloma received autografts followed by anti-CD3/anti-CD28–costimulated autologous T cells, accompanied by MAGE-A3 peptide immunizations before T-cell collection and five times after ASCT. Immune responses to the vaccine were evaluated by cytokine production (all patients), dextramer binding to CD8+ T cells, and ELISA performed serially after transplant. Results: T-cell infusions were well tolerated, whereas vaccine injection site reactions occurred in >90% of patients. Two of nine patients who received montanide developed sterile abscesses; however, this did not occur in the 18 patients who did not receive montanide. Dextramer staining demonstrated MAGE-A3–specific CD8 T cells in 7 of 8 evaluable HLA-A2+ patients (88%), whereas vaccine-specific cytokine-producing T cells were generated in 19 of 25 patients (76%). Antibody responses developed in 7 of 9 patients (78%) who received montanide and only weakly in 2 of 18 patients (11%) who did not. The 2-year overall survival was 74% [95% confidence interval (CI), 54%–100%] and 2-year event-free survival was 56% (95% CI, 37%–85%). Conclusions: A high frequency of vaccine-specific T-cell responses were generated after transplant by combining costimulated autologous T cells with a Poly-ICLC/GM-CSF–primed MAGE-A3 vaccine. Clin Cancer Res; 20(5); 1355–65. ©2013 AACR.


Bone Marrow Transplantation | 2004

Molecular remission of CML after autotransplantation followed by adoptive transfer of costimulated autologous T cells

Aaron P. Rapoport; Bruce L. Levine; Ashraf Badros; Barry Meisenberg; Kathleen Ruehle; Asit K. Nandi; S Rollins; Sabrina Natt; Bashi Ratterree; Sandra Westphal; Dean L. Mann; Carl H. June

Summary:Four patients with chronic myelogenous leukemia (CML) that was refractory to interferon alpha (two patients) or imatinib mesylate (two patients), and who lacked donors for allogeneic stem cell transplantation, received autotransplants followed by infusions of ex vivo costimulated autologous T cells. At day +30 (about 14 days after T-cell infusion), the mean CD4+ cell count was 481 cells/μl (range 270–834) and the mean CD8+ count was 516 cells/μl (range 173–1261). One patient had a relative lymphocytosis at 3.5 months after T-cell infusion, with CD4 and CD8 levels of 750 and 1985 cells/μl, respectively. All the four patients had complete cytogenetic remissions early after transplantation, three of whom also became PCR negative for the bcr/abl fusion mRNA. One patient, who had experienced progressive CML while on interferon alpha therapy, became PCR− post transplant, and remained in a molecular CR at 3.0 years of follow-up. All the four patients survived at 6, 9, 40, and 44 months post transplant; the patient who remained PCR+ had a cytogenetic and hematologic relapse of CML, but entered a molecular remission on imatinib. Autotransplantation followed by costimulated autologous T cells is feasible for patients with chronic phase CML, who lack allogeneic donors and can be associated with molecular remissions.


Bone Marrow Transplantation | 2016

Major clinical response to nivolumab in relapsed/refractory Hodgkin lymphoma after allogeneic stem cell transplantation

Jean Yared; N Hardy; Z Singh; S Hajj; Ashraf Badros; M Kocoglu; S Yanovich; E A Sausville; C Ujjani; Kathleen Ruehle; C Goecke; M Landau; Aaron P. Rapoport

Major clinical response to nivolumab in relapsed/refractory Hodgkin lymphoma after allogeneic stem cell transplantation


Bone Marrow Transplantation | 2004

Autologous stem cell transplantation followed by consolidation chemotherapy for relapsed or refractory Hodgkin's lymphoma

Aaron P. Rapoport; Chuanfa Guo; Ashraf Badros; R Hakimian; Gorgun Akpek; E Kiggundu; Barry Meisenberg; H Mannuel; Naoko Takebe; Robert G. Fenton; Javier Bolaños-Meade; Meyer R. Heyman; Ivana Gojo; Kathleen Ruehle; Sabrina Natt; Bashi Ratterree; T Withers; Clarence Sarkodee-Adoo; G Phillips; Guido Tricot

Summary:Relapse remains a major cause of treatment failure after autotransplantation (auto-PBSCT) for Hodgkins disease (HD). The administration of non-crossresistant therapies during the post-transplant period may delay or prevent relapse. We prospectively studied the role of consolidation chemotherapy (CC) after auto-PBSCT in 37 patients with relapsed or refractory HD. Patients received high-dose gemcitabine–BCNU–melphalan and auto-PBSCT followed by involved-field radiation and up to four cycles of the DCEP-G regimen, which consisted of dexamethasone, cyclophosphamide, etoposide, cisplatin, gemcitabine given at 3 and 9 months post transplant alternating with a second regimen (DPP) of dexamethasone, cisplatin, paclitaxel at 6 and 12 months post transplant. The probabilities of event-free survival (EFS) and overall survival (OS) at 2.5 years were 59% (95% CI=42–76%) and 86% (95% CI=71–99%), respectively. In all, 17 patients received 54 courses of CC and 15 were surviving event free (2.5 years, EFS=87%). There were no treatment-related deaths during or after the CC phase. Post-transplant CC is feasible and well tolerated. The impact of this approach on EFS should be evaluated in a larger, randomized study.


Bone Marrow Transplantation | 2006

Autologous stem cell transplantation followed by consolidation chemotherapy for patients with multiple myeloma

I Gojo; Barry Meisenberg; Chuanfa Guo; A Fassas; A. Murthy; R Fenton; N Takebe; Melvin B. Heyman; G L Philips; Michele Cottler-Fox; C Sarkodee-Adoo; Kathleen Ruehle; T French; M Tan; Guido Tricot; Aaron P. Rapoport

Although high-dose therapy and autologous stem cell transplant (ASCT) is superior to conventional chemotherapy for treatment of myeloma, most patients relapse and the time to relapse depends upon the initial prognostic factors. The administration of non-cross-resistant chemotherapies during the post-transplant period may delay or prevent relapse. We prospectively studied the role of consolidation chemotherapy (CC) after single autologous peripheral blood stem cell transplant (auto-PBSCT) in 103 mostly newly diagnosed myeloma patients (67 patients were ⩽6 months from the initial treatment). Patients received conditioning with BCNU, melphalan±gemcitabine and auto-PBSCT followed by two cycles of the DCEP±G regimen (dexamethasone, cyclophosphamide, etoposide, cisplatin±gemcitabine) at 3 and 9 months post-transplant and alternating with two cycles of DPP regimen (dexamethasone, cisplatin, paclitaxel) at 6 and 12 months post-transplant. With a median follow-up of 61.2 months, the median event-free survival (EFS) and overall survival (OS) are 26 and 54.1 months, respectively. The 5-year EFS and OS are 23.1 and 42.5%, respectively. Overall, 51 (49.5%) patients finished all CC, suggesting that a major limitation of this approach is an inability to deliver all planned treatments. In order to improve results following autotransplantation, novel agents or immunologic approaches should be studied in the post-transplant setting.


Clinical Lymphoma, Myeloma & Leukemia | 2006

Phase I Trial of First-Line Bortezomib/Thalidomide plus Chemotherapy for Induction and Stem Cell Mobilization in Patients with Multiple Myeloma

Ashraf Badros; Olga Goloubeva; Robert G. Fenton; Aaron P. Rapoport; Gorgun Akpek; Carolynn Harris; Kathleen Ruehle; Sandra Westphal; Barry Meisenberg


Blood | 2005

Phase I Trial of Bortezomib (V) in Combination with “DT-PACE”: Toxicity, Stem Cell Collection and Engraftment in Newly Diagnosed Multiple Myeloma (MM) Patients (Pts).

Ashraf Badros; Aaron P. Rapoport; Olga Goloubeva; Kathleen Ruehle; Sandra Westphal; S. Hefner; Barry Meisenberg


Cancer | 2015

Disparities in black and white patients with multiple myeloma referred for autologous hematopoietic transplantation: A single center study

Vishal Bhatnagar; Yin Wu; Olga Goloubeva; Kathleen Ruehle; Todd Milliron; Carolynn Harris; Aaron P. Rapoport; Saul Yanovich; Edward A. Sausville; Maria R. Baer; Ashraf Badros

Collaboration


Dive into the Kathleen Ruehle's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gorgun Akpek

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruce L. Levine

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl H. June

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge