Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cancer Res Commun ; 4(1): 200-212, 2024 01 25.
Article in English | MEDLINE | ID: mdl-38181044

ABSTRACT

Sotigalimab is an agonistic anti-CD40 mAb that can modulate antitumor immune responses. In a phase II clinical trial of sotigalimab combined with neoadjuvant chemoradiation (CRT) in locally advanced esophageal/gastroesophageal junction (E/GEJ) cancer with the primary outcome of efficacy as measured by pathologic complete response (pCR) rate, the combination induced pCR in 38% of treated patients. We investigated the mechanism of action of sotigalimab in samples obtained from this clinical trial. Tumor biopsies and peripheral blood samples were collected at baseline, following an initial dose of sotigalimab, and at the time of surgery after CRT completion from six patients. High dimensional single-cell techniques were used, including combined single-cell RNA-sequencing and proteomics (CITEseq) and multiplexed ion beam imaging, to analyze immune responses. Sotigalimab dramatically remodeled the immune compartment in the periphery and within the tumor microenvironment (TME), increasing expression of molecules related to antigen processing and presentation and altering metabolic pathways in myeloid cells. Concomitant with these changes in myeloid cells, sotigalimab treatment primed new T cell clonotypes and increased the density and activation of T cells with enhanced cytotoxic function. Sotigalimab treatment also induced a decrease in the frequency of Tregs in the TME. These findings indicate that a single dose of sotigalimab leads to enhanced antigen presentation that can activate T cells and induce new T cell clones. This restructuring of the TME provides elements which are critical to the development of effective antitumor immune responses and improved clinical outcomes.


Subject(s)
Adenocarcinoma , Antineoplastic Agents , Esophageal Neoplasms , Humans , Neoadjuvant Therapy/methods , Tumor Microenvironment , Antineoplastic Agents/therapeutic use , Esophageal Neoplasms/drug therapy
2.
Clin Cancer Res ; 30(1): 74-81, 2024 01 05.
Article in English | MEDLINE | ID: mdl-37535056

ABSTRACT

PURPOSE: Disease progression during or after anti-PD-1-based treatment is common in advanced melanoma. Sotigalimab is a CD40 agonist antibody with a unique epitope specificity and Fc receptor binding profile optimized for activation of CD40-expressing antigen-presenting cells. Preclinical data indicated that CD40 agonists combined with anti-PD1 could overcome resistance to anti-PD-1. PATIENTS AND METHODS: We conducted a multicenter, open-label, phase II trial to evaluate the combination of sotigalimab 0.3 mg/kg and nivolumab 360 mg every 3 weeks in patients with advanced melanoma following confirmed disease progression on a PD-1 inhibitor. The primary objective was to determine the objective response rate (ORR). RESULTS: Thirty-eight subjects were enrolled and evaluable for safety. Thirty-three were evaluable for activity. Five confirmed partial responses (PR) were observed for an ORR of 15%. Two PRs are ongoing at 45.9+ and 26+ months, whereas the other three responders relapsed at 41.1, 18.7, and 18.4 months. The median duration of response was at least 26 months. Two additional patients had stable disease for >6 months. Thirty-four patients (89%) experienced at least one adverse event (AE), and 13% experienced a grade 3 AE related to sotigalimab. The most common AEs were pyrexia, chills, nausea, fatigue, pruritus, elevated liver function, rash, vomiting, headache, arthralgia, asthenia, myalgia, and diarrhea. There were no treatment-related SAEs, deaths, or discontinuation of sotigalimab due to AEs. CONCLUSIONS: Sotigalimab plus nivolumab had a favorable safety profile consistent with the toxicity profiles of each agent. The combination resulted in durable and prolonged responses in a subset of patients with anti-PD-1-resistant melanoma, warranting further evaluation in this setting. See related commentary by Wu and Luke, p. 9.


Subject(s)
Melanoma , Nivolumab , Humans , Nivolumab/adverse effects , Melanoma/pathology , Antibodies, Monoclonal/adverse effects , Disease Progression , Antineoplastic Combined Chemotherapy Protocols/adverse effects
3.
JAMA Oncol ; 9(12): 1660-1668, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37824131

ABSTRACT

Importance: Metastatic soft tissue sarcomas (STSs) have limited systemic therapy options, and immunomodulation has not yet meaningfully improved outcomes. Intratumoral (IT) injection of the toll-like receptor 4 (TLR4) agonist glycopyranosyl lipid A in stable-emulsion formulation (GLA-SE) has been studied as immunotherapy in other contexts. Objective: To evaluate the safety, efficacy, and immunomodulatory effects of IT GLA-SE with concurrent radiotherapy in patients with metastatic STS with injectable lesions. Design, Setting, and Participants: This phase 1 nonrandomized controlled trial of patients with STS was performed at a single academic sarcoma specialty center from November 17, 2014, to March 16, 2016. Data analysis was performed from August 2016 to September 2022. Interventions: Two doses of IT GLA-SE (5 µg and 10 µg for 8 weekly doses) were tested for safety in combination with concurrent radiotherapy of the injected lesion. Main Outcomes and Measures: Primary end points were safety and tolerability. Secondary and exploratory end points included local response rates as well as measurement of antitumor immunity with immunohistochemistry and T-cell receptor (TCR) sequencing of tumor-infiltrating and circulating lymphocytes. Results: Twelve patients (median [range] age, 65 [34-78] years; 8 [67%] female) were treated across the 2 dose cohorts. Intratumoral GLA-SE was well tolerated, with only 1 patient (8%) experiencing a grade 2 adverse event. All patients achieved local control of the injected lesion after 8 doses, with 1 patient having complete regression (mean regression, -25%; range, -100% to 4%). In patients with durable local response, there were detectable increases in tumor-infiltrating lymphocytes. In 1 patient (target lesion -39% at 259 days of follow-up), TCR sequencing revealed expansion of preexisting and de novo clonotypes, with convergence of numerous rearrangements coding for the same binding sequence (suggestive of clonal convergence to antitumor targets). Single-cell sequencing identified these same expanded TCR clones in peripheral blood after treatment; these T cells had markedly enhanced Tbet expression, suggesting TH1 phenotype. Conclusions and Relevance: In this nonrandomized controlled trial, IT GLA-SE with concurrent radiotherapy was well tolerated and provided more durable local control than radiotherapy alone. Patients with durable local response demonstrated enhanced IT T-cell clonal expansion, with matched expansion of these clonotypes in the circulation. Additional studies evaluating synergism of IT GLA-SE and radiotherapy with systemic immune modulation are warranted. Trial Registration: ClinicalTrials.gov Identifier: NCT02180698.


Subject(s)
Sarcoma , Soft Tissue Neoplasms , Humans , Female , Aged , Male , Toll-Like Receptor 4/agonists , T-Lymphocytes , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/radiotherapy , Sarcoma/drug therapy , Sarcoma/radiotherapy , Receptors, Antigen, T-Cell
4.
Leuk Lymphoma ; 63(4): 821-833, 2022 04.
Article in English | MEDLINE | ID: mdl-34865586

ABSTRACT

Intratumoral injection of G100, a toll-like receptor 4 (TLR4) agonist, was shown pre-clinically to stimulate anti-tumor immune responses and tumor regression. This open-label, multicenter, phase 1/2 trial evaluated the safety, tolerability, and preliminary efficacy of intratumoral G100 injections following localized low-dose radiation in patients with follicular lymphoma (ClinicalTrials.gov #NCT02501473). The study was comprised of a G100 dose escalation (5 or 10 µg/dose, or 20 µg/dose for large tumors); a randomized component comparing G100 to G100 plus pembrolizumab; and G100 20 µg/dose expansion. Adverse events grade ≥3 were uncommon in patients treated with G100, and no unexpected toxicities were observed when combined with pembrolizumab. G100 20 µg (n = 18) resulted in an overall response rate of 33.3% and abscopal tumor regression in 72.2% of patients. This early-phase study provides a foundation for combining an intratumoral TLR4 agonist with agents to produce immune-mediated responses in follicular lymphoma with limited added toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Lymphoma, Follicular , Toll-Like Receptor 4 , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Humans , Lymphoma, Follicular/drug therapy , Toll-Like Receptor 4/agonists
5.
Clin Cancer Res ; 25(19): 5808-5817, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31227504

ABSTRACT

PURPOSE: LV305 is a modified, third-generation, nonreplicating, integration-deficient lentivirus-based vector designed to selectively transduce dendritic cells in vivo. LV305 induces expression of the New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1) cancer testis antigen in dendritic cells, promoting immune responses against NY-ESO-1-expressing tumors. This phase I study evaluated the safety, immunogenicity, and preliminary efficacy of LV305 in patients with sarcoma or other solid tumors. PATIENTS AND METHODS: Adults with previously treated, advanced, NY-ESO-1-positive solid tumors and limited tumor burden were eligible. LV305 was administered every 3 weeks by intradermal injection in four dose cohorts (Cohort 1: 108 vector genomes (vg) x 3 doses; Cohorts 1A, 2, and 3: 108 vg, 109 vg, 1010 vg x 4 doses). RESULTS: Thirty-nine patients were enrolled: 3 patients each in Cohorts 1, 1A, and 2, and 30 patients in Cohort 3. No dose-limiting toxicities were observed. Tumor types included sarcoma (n = 24), ovarian (n = 8), melanoma (n = 6), and lung cancer (n = 1). All treatment-related adverse events were grade 1 or 2. Common treatment-related adverse events were fatigue (49%), injection site reactions (46%), and myalgia (21%). The disease control rate was 56.4% in all patients and 62.5% in sarcoma patients. One patient with synovial sarcoma achieved a partial response lasting >36 months. Anti-NY-ESO-1-specific CD4+ and/or CD8+ T cells were induced in 57% of evaluable sarcoma patients. Induction of an anti-NY-ESO-1 immune response was associated with improved 1-year survival in an exploratory analysis. CONCLUSIONS: This first-in-class, first-in-human study of LV305 demonstrated a favorable safety profile, induction of antigen-specific responses, and potential clinical activity in patients with advanced cancer.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/transplantation , Genetic Vectors/administration & dosage , Lentivirus/genetics , Membrane Proteins/immunology , Neoplasms/therapy , Sarcoma/therapy , Adult , Aged , Antigens, Neoplasm/biosynthesis , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cohort Studies , Dendritic Cells/cytology , Dendritic Cells/immunology , Disease-Free Survival , Female , Genetic Vectors/adverse effects , Genetic Vectors/genetics , Humans , Lentivirus/immunology , Male , Membrane Proteins/biosynthesis , Middle Aged , Neoplasms/immunology , Sarcoma/immunology , Survival Rate
6.
Clin Cancer Res ; 25(4): 1185-1195, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30093453

ABSTRACT

PURPOSE: G100 is a toll-like receptor 4 (TLR4) agonist that triggers innate and adaptive antitumor immune responses in preclinical models. This pilot study assessed the safety, efficacy, and immunologic activity of intratumoral (IT) administration of G100 in patients with Merkel cell carcinoma (MCC). PATIENTS AND METHODS: Patients with locoregional MCC (n = 3; cohort A) received neoadjuvant IT G100 (2 weekly doses at 5 µg/dose) followed by surgery and radiotherapy; patients with metastatic MCC (n = 7; cohort B) received 3 doses in a 6-week cycle and could receive additional cycles with/without radiotherapy. RESULTS: IT G100 was safe and feasible in both neoadjuvant and metastatic settings. Treatment-related adverse events were mostly grade 1 or 2 injection-site reactions. IT G100 led to increased inflammation in the injected tumors with infiltration of CD8+ and CD4+ T cells and activation of immune-related genes. These proinflammatory changes were associated with local tumor regression and appeared to promote systemic immunity. All 3 cohort A patients successfully completed therapy; 2 patients remain recurrence free at 44+ and 41+ months, including 1 with a pathologic complete response after G100 alone. In cohort B, 2 patients achieved sustained partial responses, both lasting 33+ months after 2 cycles of therapy. CONCLUSIONS: In this first-in-human study, IT G100 induced antitumor immune responses, demonstrated acceptable safety, and showed encouraging clinical activity.See related commentary by Marquez-Rodas et al., p. 1127.


Subject(s)
Carcinoma, Merkel Cell/drug therapy , Immunotherapy , Lipid A/analogs & derivatives , Neoplasm Recurrence, Local/drug therapy , Toll-Like Receptor 4/agonists , Aged , Aged, 80 and over , Cancer Vaccines/administration & dosage , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Lipid A/pharmacology , Lipid A/therapeutic use , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Pilot Projects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Microenvironment/drug effects
7.
J Immunother ; 40(8): 302-306, 2017 10.
Article in English | MEDLINE | ID: mdl-28891906

ABSTRACT

Effective induction of antitumor T cells is a pivotal goal of cancer immunotherapy. To this end, lentiviral vectors (LV) are uniquely poised to directly prime CD8 T-cell responses via transduction of dendritic cells in vivo and have shown promise as active cancer therapeutics in preclinical tumor models. However, until now, significant barriers related to production and regulation have prevented their widespread use in the clinic. We developed LV305, a dendritic cell-targeting, integration-deficient, replication incompetent LV from the ZVex platform, encoding the full-length cancer-testis antigen NY-ESO-1. LV305 is currently being evaluated in phase 1 and 2 trials in metastatic recurrent cancer patients with NY-ESO-1 positive solid tumors as a single agent and in combination with anti-PD-L1. Here we report on the first patient treated with LV305, a young woman with metastatic, recurrent, therapy-refractive NY-ESO-1 synovial sarcoma. The patient developed a robust NY-ESO-1-specific CD4 and CD8 T-cell response after 3 intradermal injections with LV305, and subsequently over 85% disease regression that is continuing for >2.5 years posttherapy. No adverse events >grade 2 occurred. This case demonstrates that LV305 can be safely administered and has the potential to induce a significant clinical benefit and immunologic response in a patient with advanced stage cancer.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Lung Neoplasms/therapy , Sarcoma, Synovial/therapy , Adult , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Dendritic Cells/transplantation , Drug Resistance, Neoplasm , Female , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pneumonectomy , Programmed Cell Death 1 Receptor/metabolism , Recurrence , Remission Induction , Sarcoma, Synovial/immunology , Sarcoma, Synovial/secondary
8.
Cancer Immunol Immunother ; 64(4): 437-46, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25579378

ABSTRACT

Fresolimumab is an antibody capable of neutralizing all human isoforms of transforming growth factor beta (TGFß) and has demonstrated anticancer activity in investigational studies. Inhibition of TGFß by fresolimumab can potentially result in the development of cutaneous lesions. The aim of this study was to investigate the clinical, histological, and immunohistochemical characteristics of cutaneous neoplasms associated with fresolimumab. Skin biopsies (n = 24) were collected and analyzed from patients (n = 5) with treatment-emergent, cutaneous lesions arising during a phase 1 study of multiple doses of fresolimumab in patients (n = 29) with melanoma or renal cell carcinoma. Blinded, independent histological review and measurements of Ki-67, p53, and HPV integration were performed. Based on central review, four patients developed lesions with histological characteristics of keratoacanthomas, and of these patients, a single case of well-differentiated squamous cell carcinoma was also found. Expression of Ki-67, no evidence of p53 overexpression, and only focal positivity for human papillomavirus RNA by in situ hybridization in 4/18 cases were consistent with these findings. Following completion of fresolimumab, lesions spontaneously resolved. Therefore, benign, reversible keratoacanthomas were the most common cutaneous neoplasms observed, a finding of importance for adverse event monitoring, patient care, and optimization of therapies targeting TGFß.


Subject(s)
Antibodies, Monoclonal/adverse effects , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/chemically induced , Keratoacanthoma/chemically induced , Skin Neoplasms/chemically induced , Transforming Growth Factor beta/antagonists & inhibitors , Antibodies, Monoclonal, Humanized , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/metabolism , Humans , Keratoacanthoma/diagnosis , Keratoacanthoma/metabolism , Ki-67 Antigen/metabolism , Skin/drug effects , Skin/immunology , Skin/metabolism , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism , Transforming Growth Factor beta/immunology , Tumor Suppressor Protein p53/metabolism
9.
PLoS One ; 9(3): e90353, 2014.
Article in English | MEDLINE | ID: mdl-24618589

ABSTRACT

BACKGROUND: In advanced cancers, transforming growth factor-beta (TGFß) promotes tumor growth and metastases and suppresses host antitumor immunity. GC1008 is a human anti-TGFß monoclonal antibody that neutralizes all isoforms of TGFß. Here, the safety and activity of GC1008 was evaluated in patients with advanced malignant melanoma and renal cell carcinoma. METHODS: In this multi-center phase I trial, cohorts of patients with previously treated malignant melanoma or renal cell carcinoma received intravenous GC1008 at 0.1, 0.3, 1, 3, 10, or 15 mg/kg on days 0, 28, 42, and 56. Patients achieving at least stable disease were eligible to receive Extended Treatment consisting of 4 doses of GC1008 every 2 weeks for up to 2 additional courses. Pharmacokinetic and exploratory biomarker assessments were performed. RESULTS: Twenty-nine patients, 28 with malignant melanoma and 1 with renal cell carcinoma, were enrolled and treated, 22 in the dose-escalation part and 7 in a safety cohort expansion. No dose-limiting toxicity was observed, and the maximum dose, 15 mg/kg, was determined to be safe. The development of reversible cutaneous keratoacanthomas/squamous-cell carcinomas (4 patients) and hyperkeratosis was the major adverse event observed. One malignant melanoma patient achieved a partial response, and six had stable disease with a median progression-free survival of 24 weeks for these 7 patients (range, 16.4-44.4 weeks). CONCLUSIONS: GC1008 had no dose-limiting toxicity up to 15 mg/kg. In patients with advanced malignant melanoma and renal cell carcinoma, multiple doses of GC1008 demonstrated acceptable safety and preliminary evidence of antitumor activity, warranting further studies of single agent and combination treatments. TRIAL REGISTRATION: Clinicaltrials.gov NCT00356460.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Melanoma/drug therapy , Melanoma/pathology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/diagnosis , Female , Humans , Kidney Neoplasms/diagnosis , Male , Middle Aged , Neoplasm Staging , Positron-Emission Tomography , Tomography, X-Ray Computed , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/blood , Treatment Outcome
10.
PLoS One ; 8(6): e62785, 2013.
Article in English | MEDLINE | ID: mdl-23826077

ABSTRACT

Hypoxia and interactions with bone marrow (BM) stromal cells have emerged as essential components of the leukemic BM microenvironment in promoting leukemia cell survival and chemoresistance. High levels of transforming growth factor beta 1 (TGFß1) produced by BM stromal cells in the BM niche regulate cell proliferation, survival, and apoptosis, depending on the cellular context. Exogenous TGFß1 induced accumulation of acute myeloid leukemia (AML) cells in a quiescent G0 state, which was further facilitated by the co-culture with BM-derived mesenchymal stem cells (MSCs). In turn, TGFß-neutralizing antibody 1D11 abrogated rhTGFß1 induced cell cycle arrest. Blocking TGFß with 1D11 further enhanced cytarabine (Ara-C)-induced apoptosis of AML cells in hypoxic and in normoxic conditions. Additional constituents of BM niche, the stroma-secreted chemokine CXCL12 and its receptor CXCR4 play crucial roles in cell migration and stroma/leukemia cell interactions. Treatment with 1D11 combined with CXCR4 antagonist plerixafor and Ara-C decreased leukemia burden and prolonged survival in an in vivo leukemia model. These results indicate that blockade of TGFß by 1D11 and abrogation of CXCL12/CXCR4 signaling may enhance the efficacy of chemotherapy against AML cells in the hypoxic BM microenvironment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow/drug effects , Cytarabine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Tumor Microenvironment/drug effects , Animals , Antibodies, Neutralizing/metabolism , Benzylamines , Bone Marrow/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chemokine CXCL12/metabolism , Coculture Techniques , Cyclams , Female , Heterocyclic Compounds/pharmacology , Humans , Leukemia, Myeloid, Acute/metabolism , Mice, SCID , Neoplasm Transplantation , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Tumor Microenvironment/physiology
11.
Biol Blood Marrow Transplant ; 18(10): 1564-72, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22683613

ABSTRACT

Preapheresis peripheral blood (PB) CD34(+) cell count is a strong predictor of hematopoietic stem cell (HSC) mobilization and is routinely used to optimize the timing, cost, and success of HSC collection in patients with multiple myeloma. However, a uniform PB CD34(+) cell count that predicts mobilization failure has not been defined, resulting in the development of institute-specific algorithms for mobilization, particularly regarding the decision of when to use the novel stem cell mobilization agent plerixafor. In this post hoc analysis, we evaluated the mobilization efficacy of plerixafor plus granulocyte colony-stimulating factor (G-CSF) versus placebo plus G-CSF in patients with multiple myeloma, stratified by preapheresis PB CD34(+) cell count: <10, <15, <20, and ≥20 cells/µL. Regardless of the PB CD34(+) cell count, the total yield of CD34(+) cells from apheresis was significantly higher in the plerixafor group than in the placebo group, and significantly more patients in the plerixafor group collected the minimum (≥2 × 10(6) cells/kg) and optimum (≥6 × 10(6) cells/kg) stem cell yields on each day of apheresis. As a corollary, the greater stem cell collection in plerixafor-treated patients resulted in the need for significantly fewer days of apheresis to reach minimum and optimum cell doses across all cell count groups. For all CD34(+) cell count groups, the proportion of patients proceeding to transplantation and the median time to platelet and neutrophil engraftment were similar in the plerixafor and placebo groups. Our findings demonstrate that in patients with multiple myeloma who might be predicted to fail mobilization based on low PB CD34(+) cell count, the addition of plerixafor to G-CSF allows for collection of the minimal and optimal cell doses in a greater proportion of patients compared with G-CSF alone. In addition, plerixafor plus G-CSF significantly improves the likelihood of optimal HSC collection in patients with higher preapheresis PB CD34(+) cell counts (≥20 cells/µL) compared with placebo plus G-CSF. Collectively, this analysis of predicted poor mobilizers validates the superiority of plerixafor plus G-CSF compared with G-CSF alone, which had been demonstrated previously in the overall patient population.


Subject(s)
Antigens, CD34/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds/pharmacology , Immunologic Factors/pharmacology , Adolescent , Adult , Aged , Antigens, CD34/metabolism , Benzylamines , Biomarkers/metabolism , Blood Component Removal , Cyclams , Double-Blind Method , Drug Combinations , Female , Humans , Lymphocyte Count , Male , Middle Aged , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Placebos , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/immunology , Transplantation, Autologous
12.
Blood ; 118(18): 4963-6, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21835955

ABSTRACT

Mutations in CXCR4 cause severe leukopenia in myelokathexis or WHIM syndrome. Plerixafor inhibits binding of CXCR4 to its ligand CXCL12. We investigated the effects of plerixafor (0.04 to 0.24 mg/kg) administered at 2-4 day intervals in 6 patients. Outcome measures were the patients' complete blood cell counts, CD34(+) cell counts and lymphocyte subtypes compared with 5 normal subjects similarly treated with plerixafor. All patients showed prompt leukocytosis with maximum blood neutrophils and lymphocytes at 6-12 hours. Blood neutrophils peaked at 6-12 hours, increasing from a mean baseline of 0.4 ± 0.1 × 109/L, to mean peak of 4.5 ± 0.78 × 109/L. Lymphocytes also increased; the greatest increase was in B cells (CD19(+) cells), a > 40-fold increase over baseline at the 0.08 mg/kg dose. None of the patients experienced any significant adverse effects. Plerixafor is a promising therapy for this condition.


Subject(s)
Granulocyte Precursor Cells/drug effects , Heterocyclic Compounds/therapeutic use , Immunologic Deficiency Syndromes/drug therapy , Warts/drug therapy , Adult , Aged , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Benzylamines , Bone Marrow Diseases/drug therapy , Bone Marrow Diseases/pathology , Cyclams , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Granulocyte Precursor Cells/pathology , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/adverse effects , Heterocyclic Compounds/pharmacokinetics , Humans , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Leukopenia/complications , Leukopenia/drug therapy , Leukopenia/pathology , Male , Middle Aged , Primary Immunodeficiency Diseases , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Warts/blood , Warts/genetics , Warts/pathology
14.
Biol Blood Marrow Transplant ; 17(7): 943-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20971201

ABSTRACT

Autologous transplantation of peripheral blood (PB) hematopoietic stem cells (HSCs) is a widely used strategy for reconstitution of blood cells following high-dose chemotherapy for hematologic malignancies such as multiple myeloma (MM), non-Hodgkin lymphoma (NHL), and acute myeloid leukemia (AML), among others. Stem cells for transplantation are usually obtained from PB after treatment with chemotherapy with or without cytokine, usually granulocyte-colony stimulating factor (G-CSF), or after treatment with cytokine alone. The use of autologous peripheral blood stem cells (PBSCs) for transplantation is associated with the risk of contamination of the graft with tumor cells; whether this impacts response rates, progression-free survival (PFS), and overall survival (OS) is still debatable. This review summarizes the controversy surrounding tumor cell mobilization (TCM), the complexity of detection of minimal residual diseases, the available diagnostic tools, differences in TCM with available mobilization regimens, and the potential effect of TCM on clinical outcome. Collectively, these data suggest that new treatment paradigms to manage hematologic malignancies, such as MM, NHL, and AML, are needed and should focus on increasing the chemosensitivity of the tumor and eliminating residual disease.


Subject(s)
Cytokines/adverse effects , Granulocyte Colony-Stimulating Factor/adverse effects , Hematologic Neoplasms/pathology , Hematopoietic Stem Cell Mobilization/adverse effects , Heterocyclic Compounds/adverse effects , Neoplastic Cells, Circulating , Neoplastic Stem Cells/drug effects , Peripheral Blood Stem Cell Transplantation/adverse effects , Transplantation, Autologous/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzylamines , Biomarkers, Tumor , Combined Modality Therapy , Cyclams , Cytokines/pharmacology , DNA, Neoplasm/blood , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , Hematologic Neoplasms/blood , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/mortality , Hematologic Neoplasms/surgery , Heterocyclic Compounds/pharmacology , Humans , Neoplasm, Residual , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/cytology , Polymerase Chain Reaction/methods , Recurrence , Risk , Translocation, Genetic , Treatment Outcome
15.
Biol Blood Marrow Transplant ; 17(8): 1146-53, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21126595

ABSTRACT

Autologous hematopoietic stem cell transplantation (ASCT) is an established treatment for patients with hematologic malignancies, yet the impact of transplanted CD34(+) cell dose on clinical outcomes is unresolved. We conducted post hoc analyses of transplanted CD34(+) cell dose and hematopoietic recovery following ASCT in 438 patients with non-Hodgkin lymphoma (NHL) or multiple myeloma (MM), using data from 2 multicenter phase 3 clinical studies that compared plerixafor plus granulocyte-colony stimulating factor (G-CSF) versus placebo plus G-CSF as stem cell mobilization regimens. Days to engraftment and the proportion of patients who reached predetermined blood count thresholds were compared across 3 CD34(+) cell dose levels: 2-4 × 10(6) cells/kg, 4-6 × 10(6) cells/kg, and >6 × 10(6) cells/kg, regardless of mobilization treatment. Short-term neutrophil and platelet engraftment times were similar regardless of cell dose. A significant linear trend was observed between transplanted CD34(+) cell dose and the proportion of patients with platelet count >150 × 10(9)/L at 100 days (P < .001), 6 months (P = .026), and 12 months (P = .020) in patients with NHL, and at 100 days in patients with MM (P = .004). A linear trend was also observed between transplanted cell dose and the proportion of patients with platelet count >100 × 10(9)/L at 100 days (P < .001) and 6 months (P = .023) in patients with NHL. A higher cell dose was associated with a lower percentage of NHL patients requiring red blood cell transfusions (P = .006). Our analyses confirm previous findings that transplanted CD34(+) cell dose may be associated with better long-term platelet recovery after ASCT.


Subject(s)
Antigens, CD34/immunology , Blood Platelets/immunology , Granulocyte Colony-Stimulating Factor/therapeutic use , Heterocyclic Compounds/therapeutic use , Lymphoma, Non-Hodgkin/therapy , Multiple Myeloma/therapy , Peripheral Blood Stem Cell Transplantation/methods , Benzylamines , Cyclams , Female , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Lymphoma, Non-Hodgkin/blood , Lymphoma, Non-Hodgkin/immunology , Male , Multiple Myeloma/blood , Multiple Myeloma/immunology , Platelet Count , Survival Analysis , Transplantation, Autologous
16.
Clin Cancer Res ; 10(23): 7792-8, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15585610

ABSTRACT

PURPOSE: A multicenter, randomized study was undertaken to estimate the single agent activity of Tositumomab and to determine the contribution of radioisotope-labeling with (131)I to activity and toxicity by comparing treatment outcomes for Tositumomab and Iodine I 131 Tositumomab (BEXXAR) to an equivalent total dose of unlabeled Tositumomab. EXPERIMENTAL DESIGN: Seventy-eight patients with refractory/relapsed non-Hodgkin's lymphoma were randomized to either unlabeled Tositumomab or Iodine I 131 Tositumomab. Patients progressing after unlabeled Tositumomab could cross over to receive Iodine I 131 Tositumomab. The median follow-up at analysis was 42.6 months (range 1.9 to 71.5 months). RESULTS: Responses in the Iodine I 131 Tositumomab versus unlabeled Tositumomab groups: overall response 55% versus 19% (P = 0.002); complete response 33% versus 8% (P = 0.012); median duration of overall response not reached versus 28.1 months (95% confidence interval: 7.6, not reached); median duration of complete response not reached in either arm; and median TTP 6.3 versus 5.5 months (P = 0.031), respectively. Of the patients who had a complete response after initial Iodine I 131 Tositumomab therapy, 71% (10 of 14) continued in complete response at 29.8 to 71.1 months. Two patients who achieved a complete response after unlabeled Tositumomab had ongoing responses at 48.1 to 56.9 months. Nineteen patients received Iodine I 131 Tositumomab crossover therapy. Responses after crossover versus prior response to unlabeled Tositumomab were as follows: complete response rates of 42% versus 0% (P = 0.008); overall response 68% versus 16% (P = 0.002); median durations of overall response 12.6 versus 7.6 months (P = 0.001); and median TTP 12.4 versus 5.5 months (P = 0.01), respectively. Hematologic toxicity was more severe and nonhematologic adverse events were more frequent after Iodine I 131 Tositumomab than after Tositumomab alone. Elevated thyrotropin occurred in 5% of patients. Seroconversion to human antimurine antibody after Iodine I 131 Tositumomab, unlabeled Tositumomab, and Iodine I 131 Tositumomab-crossover was 27%, 19%, and 0%, respectively. CONCLUSIONS: Unlabeled Tositumomab showed single agent activity, but in this direct comparison, all of the therapeutic outcome measures were significantly enhanced by the conjugation of (131)I to Tositumomab.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Iodine Radioisotopes/therapeutic use , Lymphoma, B-Cell/radiotherapy , Neoplasm Recurrence, Local/radiotherapy , Radioimmunotherapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/radiotherapy , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Remission Induction
17.
J Clin Oncol ; 22(23): 4717-24, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15483014

ABSTRACT

PURPOSE: The unique immunoglobulin idiotype (Id) expressed by each B-cell lymphoma is a target for immunotherapy. Vaccination with Id induces humoral and/or cellular anti-Id immune responses. However, the clinical impact of these anti-Id immune responses is unknown. We and others have previously reported that immunoglobulin G Fc receptor (FcgammaR) polymorphisms predict the clinical response of lymphoma patients to passive anti-CD20 antibody infusions. In this study, we tested whether anti-Id immune responses or FcgammaR polymorphisms associate with clinical outcome of patients who received Id vaccination. PATIENTS AND METHODS: We analyzed 136 patients with follicular lymphoma who had received Id vaccination. The anti-Id immune responses were measured and FcgammaRIIIa and FcgammaRIIa polymorphisms were determined and correlated with clinical outcome for these patients. RESULTS: Patients who mounted humoral immune responses had a longer progression-free survival (PFS) than those who did not (8.21 v 3.38 years; P = .018). Patients with FcgammaRIIIa 158 valine/valine (V/V) genotype also had a longer PFS than those with valine/phenylalanine (V/F) or phenylalanine/phenylalanine (F/F) genotypes (V/V, 8.21 v V/F, 3.38 years; P = .004; v F/F, 4.47 years; P = .035). Multivariate analysis using the Cox proportional hazards model showed that V/V genotype and humoral immune responses were independent positive predictors for PFS. CONCLUSION: This study is the first to identify the predictive value of FcgammaR polymorphism on clinical outcome in patients who received active immunotherapy with tumor antigen vaccines. Our results imply that the antibodies induced against a tumor antigen are beneficial and that FcgammaR-bearing cells mediate an antitumor effect by killing antibody-coated tumor cells.


Subject(s)
Antigens, CD/metabolism , Immunoglobulin Idiotypes/therapeutic use , Lymphoma, Non-Hodgkin/therapy , Polymorphism, Genetic , Receptors, IgG/metabolism , Antigens, CD/genetics , Cohort Studies , Female , Genetic Markers , Humans , Immunotherapy, Active/methods , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/therapy , Lymphoma, Follicular/genetics , Lymphoma, Follicular/immunology , Lymphoma, Follicular/mortality , Lymphoma, Follicular/therapy , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/mortality , Male , Multivariate Analysis , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Prospective Studies , Receptors, IgG/genetics , Risk Assessment , Sensitivity and Specificity , Survival Analysis , Treatment Outcome , Vaccination/methods
18.
J Immunother ; 25(6): 455-68, 2002.
Article in English | MEDLINE | ID: mdl-12439343

ABSTRACT

CTLA4 is a negative regulator of the costimulatory signals induced by the interaction of CD28 on T cells and B7 on dendritic cells (DCs). Antibodies (Abs) against CTLA4 can block its function and increase the activation of T cells primed to recognize antigens. The effect of CTLA4 blockade on the cross-presentation of tumor antigens by DCs to T cells was examined. Immune T cells and DC precursors were collected from patients receiving idiotype protein-pulsed DC vaccines, exposed to antigen, and examined for antitumor activity by measuring intracellular cytokine production by FACS. Idiotype-specific activation occurred in CD8+ and CD4+ T-cell populations and was up to 58 fold higher with CTLA4 blockade. These T cells could be expanded quickly and maintained tumor cytolytic activity. T-cell responses to whole tumor cell-pulsed DCs were then examined. DCs contain Fc receptors and efficiently phagocytose lymphoma cells when coated with opsonizing anti-CD20 Abs. Within a few hours, DCs ingested tumor cells and labeled proteins were observed in the cytoplasm. When anti-CD20 Ab-coated tumor-pulsed DCs were used in combination with CTLA4 blockade, up to 15 fold higher activation of Id-specific CD8+ and 3 fold higher CD4+ T cells resulted. Thus, CTLA4 blockade can enhance the measurement of Ag-specific T-cell responses and the expansion of T cells for clinical studies. In addition, the combination of CTLA4 blockade and Ab targeting of tumor to DCs is an effective method for the cross-presentation of tumor cell antigens.


Subject(s)
Antigen Presentation , Antigens, CD20/immunology , Antigens, Differentiation/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Immunoconjugates , Immunoglobulin Idiotypes/immunology , Lymphocyte Activation/drug effects , Lymphoma, Follicular/immunology , Neoplastic Stem Cells/immunology , Opsonin Proteins/immunology , T-Lymphocyte Subsets/immunology , Abatacept , Antigens, CD , B7-1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen , Cancer Vaccines , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Humans , Lymphoma, Follicular/chemistry , Lymphoma, Follicular/pathology , Neoplastic Stem Cells/chemistry , Phagocytosis , T-Lymphocyte Subsets/metabolism
19.
Blood ; 99(5): 1517-26, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11861263

ABSTRACT

Tumor-specific clonal immunoglobulin expressed by B-cell lymphomas (idiotype [Id]) can serve as a target for active immunotherapy. We have previously described the vaccination of 4 patients with follicular lymphoma using dendritic cells (DCs) pulsed with tumor-derived Id protein and now report on 35 patients treated using this approach. Among 10 initial patients with measurable lymphoma, 8 mounted T-cell proliferative anti-Id responses, and 4 had clinical responses--2 complete responses (CRs) (progression-free [PF] for 44 and 57 months after vaccination), 1 partial response (PR) (PF for 12 months), and 1 molecular response (PF for 75+ months). Subsequently, 25 additional patients were vaccinated after first chemotherapy, and 15 of 23 (65%) who completed the vaccination schedule mounted T-cell or humoral anti-Id responses. Induction of high-titer immunoglobulin G anti-Id antibodies required coupling of Id to the immunogenic carrier protein keyhole limpet hemocyanin (Id-KLH). These antibodies could bind to and induce tyrosine phosphorylation in autologous tumor cells. Among 18 patients with residual tumor at the time of vaccination, 4 (22%) had tumor regression, and 16 of 23 patients (70%) remain without tumor progression at a median of 43 months after chemotherapy. Six patients with disease progression after primary DC vaccination received booster injections of Id-KLH protein, and tumor regression was observed in 3 of them (2 CRs and 1 PR). We conclude that Id-pulsed DC vaccination can induce T-cell and humoral anti-Id immune responses and durable tumor regression. Subsequent boosting with Id-KLH can lead to tumor regression despite apparent resistance to the primary DC vaccine.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/transplantation , Immunoglobulin Idiotypes/immunology , Lymphoma, B-Cell/therapy , Adult , Aged , Antibodies, Anti-Idiotypic/biosynthesis , Antibody Formation , Cancer Vaccines/immunology , Dendritic Cells/immunology , Follow-Up Studies , Hemocyanins/administration & dosage , Hemocyanins/immunology , Humans , Immunoglobulin Idiotypes/therapeutic use , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/immunology , Male , Middle Aged , T-Lymphocytes/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...