Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Microb Cell Fact ; 23(1): 195, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971787

ABSTRACT

This study explores a sustainable approach for synthesizing silver nanocomposites (AgNCs) with enhanced antimicrobial and bioactivity using safe Lactobacillus strains and a whey-based medium (WBM). WBM effectively supported the growth of Lactobacillus delbrueckii and Lactobacillus acidophilus, triggering a stress response that led to AgNCs formation. The synthesized AgNCs were characterized using advanced spectroscopic and imaging techniques such as UV‒visible, Fourier transform infrared (FT-IR) spectroscopy, transmission electron (TEM), and scanning electron microscopy with energy dispersive X-ray analysis (SEM-Edx). Lb acidophilus-synthesized AgNCs in WBM (had DLS size average 817.2-974.3 ± PDI = 0.441 nm with an average of metal core size 13.32 ± 3.55 nm) exhibited significant antimicrobial activity against a broad spectrum of pathogens, including bacteria such as Escherichia coli (16.47 ± 2.19 nm), Bacillus cereus (15.31 ± 0.43 nm), Clostridium perfringens (25.95 ± 0.03 mm), Enterococcus faecalis (32.34 ± 0.07 mm), Listeria monocytogenes (23.33 ± 0.05 mm), methicillin-resistant Staphylococcus aureus (MRSA) (13.20 ± 1.76 mm), and filamentous fungi such as Aspergillus brasiliensis (33.46 ± 0.01 mm). In addition, Lb acidophilus-synthesized AgNCs in WBM exhibit remarkable free radical scavenging abilities, suggesting their potential as bioavailable antioxidants. These findings highlight the dual functionality of these biogenic AgNCs, making them promising candidates for applications in both medicine and nutrition.


Subject(s)
Microbial Sensitivity Tests , Nanocomposites , Silver , Whey , Nanocomposites/chemistry , Silver/chemistry , Silver/pharmacology , Whey/chemistry , Whey/metabolism , Lactobacillus acidophilus/drug effects , Lactobacillus acidophilus/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/biosynthesis , Metal Nanoparticles/chemistry , Lactobacillus/metabolism , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Spectroscopy, Fourier Transform Infrared
2.
Ann Oncol ; 20(6): 1068-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19188136

ABSTRACT

BACKGROUND: This trial investigated the efficacy and safety of weekly cetuximab combined with two different schedules of paclitaxel/carboplatin for stage IIIB/IV non-small-cell lung cancer (NSCLC). METHODS: A total of 168 patients with previously untreated stage IIIB/IV NSCLC were randomized to arm A, cetuximab (400 mg/m(2) day 1 followed by weekly 250 mg/m(2)) + paclitaxel (Taxol) (225 mg/m(2))/carboplatin (AUC6) day 1 every 3 weeks or arm B, same cetuximab regimen plus paclitaxel (100 mg/m(2)) days 1, 8, and 15 every 3 weeks and carboplatin (AUC6) day 1 every 4 weeks. Treatment continued for a four-cycle maximum. Patients with a complete response, partial response, or stable disease after four cycles could receive cetuximab 250 mg/m(2)/week until disease progression or unacceptable toxicity. The primary end point was to evaluate progression-free survival (PFS). RESULTS: Median PFS was 4.7 and 4.3 months for arms A and B, respectively (6-month PFS, 27.3% versus 30.9%). Median overall survival was 11.4 versus 9.8 months for arms A and B, respectively; estimated 1-year survival, 47.7% versus 39.3%; and objective response rate, 29.6% versus 25%. The regimen was well tolerated with rash and hematologic toxicity being most common. CONCLUSIONS: This study did not meet the prespecified benchmark of 35% 6-month PFS rate; both combination schedules of cetuximab plus paclitaxel/carboplatin were feasible and equivalent for treating advanced NSCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/pathology , Cetuximab , Dose-Response Relationship, Drug , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Paclitaxel/administration & dosage
3.
Dev Neurorehabil ; 11(1): 60-80, 2008.
Article in English | MEDLINE | ID: mdl-17943507

ABSTRACT

RATIONALE: Cerebral palsy (CP) constitutes a substantial portion of paediatric rehabilitation, yet little is known regarding actual occupational therapy (OT) and physical therapy (PT) practices. This study describes OT and PT practices for young children with CP in Quebec, Canada. METHODS: This was a cross-sectional survey. All eligible, consenting paediatric occupational therapists (OTs) and physical therapists (PTs) were interviewed using a structured telephone interview based on vignettes of two typical children with CP at two age points--18 months and 4 years. Reported practices were grouped according to the International Classification of Functioning, Disability and Health (ICF). RESULTS: 91.9% of PTs (n=62; 83.8% participation rate) and 67.1% of OTs (n=85; 91.4% participation rate) reported using at least one standardized paediatric assessment. OT and PT interventions focused primarily on impairments and primary function (such as gait function and activities of daily living). Both professions gave little attention to interventions related to play and recreation/leisure. Clinicians reported the need for more training and education specific to CP and to the use of research findings in clinical practice. CONCLUSION: Wide variations and gaps were identified in clinicians' responses suggesting the need for a basic standard of OT and PT management as well as strategies to encourage knowledge dissemination regarding current best practice.


Subject(s)
Cerebral Palsy/rehabilitation , Occupational Therapy/standards , Pediatrics/standards , Physical Therapy Modalities/standards , Quality of Health Care , Activities of Daily Living , Child, Preschool , Cross-Sectional Studies , Female , Humans , Infant , Interviews as Topic , Male , Quebec , Treatment Outcome
4.
J Clin Oncol ; 19(13): 3234-43, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11432891

ABSTRACT

PURPOSE: To evaluate the safety, pharmacokinetics, and efficacy of a chimeric anti-epidermal growth factor receptor monoclonal antibody, cetuximab, in combination with radiation therapy (RT) in patients with advanced squamous cell carcinoma of the head and neck. PATIENTS AND METHODS: We treated 16 patients in five successive treatment schedules. A standard dose escalation procedure was used; three patients entered onto the study at each dose level of cetuximab received conventional RT (70 Gy, 2 Gy/d), and the final three patients received hyperfractionated RT (76.8 Gy, 1.2 Gy bid). Cetuximab was delivered as a loading dose of 100 to 500 mg/m(2), followed by weekly infusions of 100 to 250 mg/m(2) for 7 to 8 weeks. Circulating levels of cetuximab during therapy were determined using a biomolecular interaction analysis core instrument. Human antichimeric antibody response was evaluated with a double-antigen radiometric assay. The recommended phase II/III dose was defined as the optimal cetuximab dose level based on the pharmacologic parameters and adverse events. RESULTS: The most commonly reported adverse events were fever, asthenia, transaminase elevation, nausea, and skin toxicities (grade 1 to 2 in most patients). Skin toxicity outside of the RT field was not strictly dose-dependent; however, grade 2 or higher events were observed in patients treated with higher dose regimens. There was one grade 4 allergic reaction. Most acute adverse effects were associated with RT (xerostomia, mucositis, and local skin toxicity). No antibodies against cetuximab were detected. All patients achieved an objective response (13 complete and two partial remissions). CONCLUSION: Cetuximab can be safely administered with RT. The recommended dose for phase II/III studies is a loading dose of 400 to 500 mg/m(2) and a maintenance weekly dose of 250 mg/m(2).


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Radiotherapy/methods , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Cetuximab , Combined Modality Therapy , Dose-Response Relationship, Drug , Female , Humans , Male , Metabolic Clearance Rate , Middle Aged
5.
Cancer Biother Radiopharm ; 16(2): 125-32, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11385959

ABSTRACT

The angiogenic response of a progressing malignancy is characterized by a shift in the balance of stimulatory and inhibiting factors of angiogenesis. Recognition of the regulated steps in tumor angiogenesis provides unique targets for developing anti-tumor therapy. Vitaxin is a humanized monoclonal antibody, which has specificity for the integrin alpha v beta 3 (vitronectin receptor). This antibody can impair the vascular response of endothelial cell growth factors in vitro and inhibit tumor cell mediated angiogenesis in pre-clinical animal models. Patients with metastatic cancer who failed standard therapy received intravenous doses of 10, 50 or 200 mg in cohorts of three patients. The unlabeled dose of Vitaxin was infused on days 0 and 21 of a treatment cycle. All patients received a pre-therapy imaging dose of 1 mg of Tc-99m Vitaxin with gamma camera imaging studies. There was no significant toxicity noted in these three dose levels. There were no objective anti-tumor responses. Three patients received two cycles of therapy and had stable disease at day 85 when taken off study. Radioimaging of tumor vasculature was unsuccessful although one patient with alpha v beta 3 positive melanoma had imaging of tumor sites. There was no immune response to Vitaxin in any patient. Patients receiving 10 mg doses of Vitaxin had poor plasma recovery of injected doses and brief circulation in plasma. Doses of 50 and 200 mg had plasma recovery that better approximated the predicted levels in plasma and circulation half-lives of approximately 7 days. This data suggests that an every three-week schedule of Vitaxin at doses of 200 mg (2.5-3.5 mg/kg) can maintain circulating levels of antibody with little or no toxicity. Future studies will be challenged to define anti-tumor activity in malignancy or appropriate surrogates of anti-tumor effect and explore escalating doses and alternate schedules of administration.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Neoplasms/drug therapy , Receptors, Vitronectin/immunology , Adolescent , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Dose-Response Relationship, Immunologic , Female , Humans , Male , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Organotechnetium Compounds , Pilot Projects , Radionuclide Imaging
6.
J Clin Oncol ; 18(21 Suppl): 47S-53S, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11060327

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFr) is overexpressed in a majority of head and neck squamous cell carcinomas, and this overexpression is associated with a poor prognosis. Therefore, EGFr has become the target of investigations aimed at disabling the receptor to determine whether this process leads to improved tumor kill with conventional treatment. MATERIALS AND METHODS: C225 is an anti-EGFr monoclonal antibody that inhibits receptor activity by blocking the ligand binding site. A panel of human head and neck squamous cell carcinoma cell lines was used to study the combination of C225 and radiation. RESULTS: It was determined that the combination of C225 (5 microgram/mL) delivered simultaneously with radiation (3 Gy) resulted in a greater decrement in cellular proliferation than either treatment alone. This reduction in proliferation correlated with reduced EGFr tyrosine phosphorylation and a reduction in phosphorylated signal transducer and activator of transcription-3 (STAT-3) protein (known to protect cells from apoptosis). Also, the decrement in proliferation correlated with increased apoptotic events, thereby indirectly linking C225/radiation-induced regulation of STAT-3 protein to apoptosis. CONCLUSION: This preclinical work serves as important support for the ongoing clinical investigation of C225 and radiotherapy for patients with head and neck carcinomas. The initial results of these clinical studies have been promising.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/therapy , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/therapy , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Clinical Trials as Topic , Combined Modality Therapy , ErbB Receptors/immunology , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/pathology , Humans , Radiography , Tumor Cells, Cultured
7.
J Clin Oncol ; 18(11): 2282-92, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10829049

ABSTRACT

PURPOSE: We conducted a phase I clinical trial of BR96-Doxorubicin (BR96-Dox), a chimeric anti-Lewis Y (Le(Y)) monoclonal antibody conjugated to doxorubicin, in patients whose tumors expressed the Le(Y) antigen. The study aimed to determine the toxicity, maximum-tolerated dose, pharmacokinetics, and immunogenicity of BR96-Dox. PATIENTS AND METHODS: This was a phase I dose escalation study. BR96-Dox was initially administered alone as a 2-hour infusion every 3 weeks. The occurrence of gastrointestinal (GI) toxicity necessitated the administration of BR96-Dox as a continuous infusion over 24 hours and use of antiemetics and antigastritis premedication. Patients experiencing severe GI toxicity underwent GI endoscopy. All patients underwent restaging after two cycles. RESULTS: A total of 66 patients predominantly with metastatic colon and breast cancer were enrolled onto the study. The most common side effects were GI toxicity, fever, and elevation of pancreatic lipase. At higher doses, BR96-Dox was associated with nausea, vomiting, and endoscopically documented exudative gastritis of the upper GI tract, which was dose-limiting at a maximum dose of 875 mg/m(2) (doxorubicin equivalent, 25 mg/m(2)) administered every 3 weeks. Toxicity was reversible and generally of short duration. Premedication with the antiemetic Kytril (granisetron hydrochloride; SmithKline Beecham, Philadelphia, PA), the antacid omeprazole, and dexamethasone was most effective in ameliorating GI toxicity. A dose of 700 mg/m(2) BR96-Dox (doxorubicin equivalent, 19 mg/m(2)) every 3 weeks was determined to be the optimal phase II dose when administered with antiemetic and antigastritis prophylaxis. BR96-Dox deposition on tumor tissue was documented immunohistochemically and by confocal microscopy. At the 550-mg/m(2) dose, the half-life (mean +/- SD) of BR96 and doxorubicin was 300 +/- 95 hours and 43 +/- 4 hours, respectively. BR96-Dox elicited a weak immune response in 37% of patients. Objective clinical responses were seen in two patients. CONCLUSION: BR96-Dox provides a unique strategy to deliver doxorubicin to Le(Y)-expressing tumor and was well tolerated at doses of 700 mg/m(2) every 3 weeks. BR96-Dox was not associated with the typical side-effect profile of native doxorubicin and can potentially deliver high doses of doxorubicin to antigen-expressing tumors. A phase II study in doxorubicin-sensitive tumors is warranted.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Tumor-Associated, Carbohydrate/metabolism , Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Immunotoxins/therapeutic use , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/metabolism , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Immunotoxins/adverse effects , Immunotoxins/pharmacokinetics , Lewis Blood Group Antigens/immunology , Male , Middle Aged , Neoplasms, Glandular and Epithelial/immunology , Treatment Outcome
8.
Semin Oncol ; 27(6 Suppl 12): 99-103, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11226008

ABSTRACT

We conducted a prospective pilot phase I/II clinical trial to evaluate the toxicity and response rate of the chimeric anti-CD20 monoclonal antibody, rituximab (Rituxan; Genentech, Inc, South San Francisco, CA, and IDEC Pharmaceutical Corporation, San Diego, CA), in the treatment of patients with immune thrombocytopenic purpura. Patients with a clinical diagnosis of idiopathic thrombocytopenic purpura who had failed corticosteroid therapy and whose platelet count was less than 75,000/microL were eligible for the study. Rituximab was administered in a dose-escalation fashion using doses ranging from 50 to 375 mg/m2 weekly for 4 weeks. Thirteen patients have been enrolled on the trial to date and 12 have completed the full course of treatment. No unusual toxicity was noted in this patient population. None of the three patients at the lowest dose level achieved a clinical response. Three of nine patients (30%) who have received rituximab at doses close or equal to the full dose have shown an objective clinical response (two complete responses, one partial response). The study is currently ongoing, and conclusions regarding the overall response rate, clinical parameters that influence response, surrogate markers of response, and the underlying mechanism of response remain to be addressed. The current study should provide answers to a number of important questions regarding the role of rituximab in the treatment of this and other autoimmune disorders.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Clinical Protocols , Combined Modality Therapy , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Pilot Projects , Purpura, Thrombocytopenic, Idiopathic/surgery , Rituximab , Splenectomy
9.
Clin Cancer Res ; 5(9): 2330-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499601

ABSTRACT

The principal objectives of this trial were twofold: (a) to examine the safety and relative efficacy of intradermal needle injection versus s.c. jet administration of a carcinoembryonic antigen (CEA)-encoding recombinant vaccinia virus (rV-CEA) over a limited dose range and (b) to evaluate CEA-specific immune responses or antitumor effects induced by rV-CEA vaccination. Patients were randomly assigned to one of two groups, depending upon the technique of vaccine administration. All 20 patients received two doses of 10(7) or 10(8) pfu of rV-CEA at a 4-week interval. Toxicity was limited to modest local inflammation at the inoculation site as well as low-grade fever and increased fatigue, each affecting fewer than 20% of the patients. No evidence of CEA-specific lymphoproliferation, interleukin 2 release, delayed-type hypersensitivity, or antibody response was observed. Thus, the efficacy comparison between the two administration techniques was based upon the induction of immune responses to the vaccinia virus vector. Both techniques induced vaccinia-specific lymphoproliferation, interleukin 2 release, and antibody responses of comparable magnitude and frequency as well as protected 80% of patients against pustule formation following vaccinia scarification. Thus, there is no compelling reason to recommend one administration technique over the other based upon toxicity or efficacy. We have selected s.c. jet injection for subsequent trials of rV-CEA based on the ability to accommodate larger injection volumes, enhanced standardization between clinicians, and avoidance of needles that could transmit the vaccine or blood-borne pathogens to health care workers. We recommend use of 10(8) pfu doses for subsequent trials of recombinant vaccinia virus vaccines based upon the favorable toxicity profile and more consistent local pustule formation indicative of an adequate inoculation of live virus. No objective clinical responses to the rV-CEA vaccine were observed among this population of patients with widely metastatic adenocarcinoma.


Subject(s)
Adenocarcinoma/secondary , Adenocarcinoma/therapy , Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/genetics , Vaccinia virus/genetics , Adenocarcinoma/immunology , Adult , Aged , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoembryonic Antigen/administration & dosage , Carcinoembryonic Antigen/biosynthesis , Carcinoembryonic Antigen/immunology , Female , Humans , Injections, Intradermal , Injections, Subcutaneous , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Male , Middle Aged , Vaccinia virus/immunology
10.
Cancer Biother Radiopharm ; 14(6): 451-63, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10850332

ABSTRACT

BACKGROUND: Monoclonal antibodies (mAb) to epidermal growth factor receptor (EGFr) inhibit tumor cell proliferation and enhance cytotoxicity of chemotherapeutic agents. The purpose of this study was to investigate the interaction of the anti-EGFr antibody C225 combined with radiotherapy (RT) on EGFr expressing A431 human epidermoid cancer cells. METHODS: Cell proliferation, apoptosis, EGFr expression and phosphorylation, and clonogenic survival were assayed in vitro. A431 tumor growth inhibition and immunohistochemistry analysis of EGFr expression and apoptosis were assessed in vivo. RESULTS: C225 plus RT produced greater inhibition of A431 cell proliferation than C225 or RT alone which was corroborated by enhanced apoptosis. Similar clonogenic survival occurred following the addition of C225 to RT, although colonies were smaller in the presence of C225. C225 produced inhibition of EGF-induced phosphorylation of EGFr without concurrent down-regulation of surface receptor, which was not altered by RT. Combined treatment of mice bearing tumors demonstrated enhancement of complete regressions, reduction in time to tumor size doubling, and prolongation of survival. Significant apoptosis occurred in xenograft tumors treated with C225 with or without RT. CONCLUSIONS: These data demonstrate an interaction between C225 and RT. C225-mediated apoptosis and inhibition of EGFr phosphorylation may be critical in the interaction. Studies to define the precise influence of combined modality treatment on the EGFr signal transduction cascade need to be pursued. The combination of growth factor receptor antibodies and RT has potential application in clinical oncology.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Animals , Antibodies, Monoclonal/toxicity , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Cell Division/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cetuximab , Combined Modality Therapy , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Mice , Mice, Nude , Phosphorylation , Transplantation, Heterologous , Tumor Cells, Cultured
11.
J Immunother ; 21(5): 379-88, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9789200

ABSTRACT

Melimmune is a dual preparation of two murine anti-idiotypic antibodies (anti-Ids), Melimmune-1 and Melimmune-2, which mimic separate epitopes of the melanoma-associated high molecular weight proteoglycan antigen. In an animal model, vaccination with either anti-Id leads to tumor rejection, and Phase I clinical trials have demonstrated the tolerance of each reagent in humans. We conducted a Phase IB trial of different doses of a one-to-one composition to Melimmune-1 and Melimmune-2 administered with SAF-m adjuvant in patients with resected melanoma without evidence of metastatic disease. A total of 21 patients were enrolled in this multicenter trial. Detailed immune response analysis was conducted on 13 patients enrolled at a single institution. Following vaccination, 12 of the 13 patients demonstrated antibodies to both Melimmune-1 and Melimmune-2, including significant anti-V-region reactivity. Maximum anti-V-region reactivity was generally detected following the last vaccination. Anti-V-region reactivity directed at Melimmune-1 and Melimmune-2 in excess of 1 microgram/ml was detected in 4 and 10 of 12 patients, respectively. Sera from patients obtained at time of peak anti-V-region reactivity did not demonstrate the ability to inhibit Ab1 binding to tumor cells or direct anti-tumor cell reactivity. However, in vitro cellular proliferation was observed in response to Melimmune-1 and/or Melimmune-2 F(Ab')2 in all patients with a mean stimulation index of 12.0 and 27.8, respectively. Overall, the antibody and cellular immune response to Melimmune-2 was more potent than to Melimmune-1, and all antibody doses elicited an immune response. The optimal biologic dose of Melimmune could not be determined in this small patient population.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Immunotherapy, Active , Melanoma/immunology , Proteoglycans/immunology , Adult , Aged , Animals , Antibodies, Neoplasm/analysis , Epitopes/immunology , Female , Humans , Immunity, Cellular/drug effects , Lymphocyte Activation/drug effects , Male , Melanoma/drug therapy , Mice , Middle Aged , Neoplasm Proteins/immunology
12.
Biotech Histochem ; 73(4): 186-97, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9735877

ABSTRACT

In this study, we compare various methods for the detection of a tumor-associated target antigen and deposition of the bound therapeutic monoclonal antibody in patients enrolled in two separate trials, one involving the administration of two radiolabeled monoclonal antibodies and the other involving an unlabeled antibody. In the first trial, patients with TAG-72 expressing metastatic colon cancer scheduled for surgical intervention received radiolabeled murine and chimeric B72.3 antibody followed by radioimmune imaging and subsequent laparotomy. Normal and tumor tissues obtained at surgery were processed for routine histology, immunohistochemistry, radiometry, and autoradiography. Both anti-TAG-72 antibodies localized to known tumor sites as evidenced by radioimmune imaging. Resected tissue revealed a high tumor-to-normal radiolocalization ratio, and autoradiography demonstrated even deposition of the radiolabeled antibodies throughout the entire tumor deposit with sparing of surrounding normal tissue. In contrast, immunohistochemistry on the same sections revealed comparatively weak antigen expression and patchy antibody localization. In the second trial, patients with GD2 antigen expressing metastatic melanoma received the unlabeled chimeric anti-GD2 antibody C14.18. Immunologic detection of the GD2 antigen and C14.18 deposition was performed on biopsy section as well as on single cell suspension. FACS analysis of the single cell suspension proved more sensitive for the detection of bound antibody than immunohistochemistry, although both methods yielded comparable results for GD2 antigen expression. Our findings demonstrate that the optimal method for the detection of tumor-associated antigen and bound therapeutic antibody can vary depending upon the nature of the antibody (radiolabeled vs. unlabeled and murine vs. chimeric), fixation stability of the target antigen, and the type of pathologic material available for study.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Colonic Neoplasms/immunology , Gangliosides/immunology , Glycoproteins/immunology , Melanoma/immunology , Animals , Antibody Specificity , Autoradiography , Clinical Trials as Topic , Colonic Neoplasms/pathology , Evaluation Studies as Topic , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Melanoma/pathology , Mice
14.
J Am Acad Dermatol ; 39(1): 63-73, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9674399

ABSTRACT

BACKGROUND: DAB389IL-2 is a novel fusion toxin that retargets the cytotoxic A-chain of diphtheria toxin to interleukin-2 (IL-2) receptor-expressing tumors. OBJECTIVE: The purpose of this phase I trial was to study the toxicity, maximum tolerated dose, and clinical efficacy of DAB389IL-2 in IL-2 receptor expressing lymphoproliferative malignancies, including cutaneous T-cell lymphoma. METHODS: DAB389IL-2 was administered intravenously daily for 5 days every 3 weeks. Dose escalation occurred between patient groups. Patients were monitored for laboratory and clinical toxicity, kinetics, immune response, and clinical efficacy. RESULTS: Thirty-five patients with cutaneous T-cell lymphoma (including 30 patients with mycosis fungoides) were treated. Previously, conventional therapy had not worked for 34 of the patients. Thirteen patients (37%) achieved an objective response, including a complete response in five patients (14%). Complete response was achieved in patients with extensive erythroderma and tumor stage mycosis fungoides. Adverse events consisted of reversible fever/chills, hypotension, nausea/vomiting, and elevation of hepatic transaminase. Doses of less than 31 microg/kg per day were well tolerated. Clinical responses were observed at all dose levels. CONCLUSION: DAB389IL-2 is well tolerated at doses of less than 31 microg/kg per day, and it induced clinical responses in previously treated mycosis fungoides, providing evidence for the antitumor activity of this molecule.


Subject(s)
Antineoplastic Agents/therapeutic use , Diphtheria Toxin/therapeutic use , Immunosuppressive Agents/therapeutic use , Immunotoxins/therapeutic use , Interleukin-2/therapeutic use , Mycosis Fungoides/therapy , Recombinant Fusion Proteins/therapeutic use , Skin Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Dermatitis, Exfoliative/therapy , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/adverse effects , Female , Fever/etiology , Humans , Hypotension/etiology , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunotoxins/administration & dosage , Immunotoxins/adverse effects , Infusions, Intravenous , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Lymphoma, T-Cell, Cutaneous/therapy , Male , Middle Aged , Mycosis Fungoides/pathology , Nausea/etiology , Neoplasm Staging , Receptors, Interleukin-2/antagonists & inhibitors , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Remission Induction , Shivering/physiology , Skin Neoplasms/pathology , Vomiting/etiology
15.
Blood ; 91(2): 399-405, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9427692

ABSTRACT

The purpose of this study was to evaluate the safety, tolerability, pharmacokinetics, and possible antitumor activity of a ligand fusion-protein, DAB389IL-2, in a phase I trial. This was a multicenter, open-label, dose-escalation trial. Patients with preserved organ function and histologically confirmed relapsed cutaneous T-cell lymphoma (CTCL), other non-Hodgkin's lymphomas (NHL), or Hodgkin's disease (HD) were eligible if their cancer was shown to express the interleukin (IL)-2 receptor by an immunohistochemical assay for the p55 or the p75 subunit. Patients received up to eight courses of DAB389IL-2 given as a short intravenous infusion daily for 5 days with subsequent courses every 21 days. The maximum tolerated dose (MTD) and tumor response was determined according to standard criteria. Seventy-three patients (44 men/29 women), aged 16 to 81 years (mean, 50.7) with CTCL (n = 35), NHL (n = 17), and HD (n = 21) were enrolled. The patients were extensively treated, failing 0 to 15 previous therapies (median, 4). Patients received one to six courses (mean, 3.3) of DAB389IL-2 over a range of 3 to 31 micrograms/kg/day. The dose-limiting toxicity was asthenia, establishing the maximum tolerated dose of 27 micrograms/kg/day. Approximately half of all patients had significant titers of antibody to diphtheria toxin or to DAB389IL-2 at the time of enrollment compared with 92% with titers at the end of treatment. The presence of antibody did not preclude clinical response. There were five complete (CR) and eight partial (PR) remissions in patients with CTCL with one CR and two PR occurring in NHL. The median time to response was 2 months and the duration of response was 2 to 39+ months. No responses were documented in patients with HD. DAB389IL-2 is well tolerated with an MTD of 27 micrograms/kg/day. This ligand fusion-protein showed antitumor effects in patients with IL-2 receptor expressing CTCL and NHL. Additional trials in these diseases are warranted.


Subject(s)
Antineoplastic Agents/therapeutic use , Diphtheria Toxin/therapeutic use , Interleukin-2/therapeutic use , Lymphoma/drug therapy , Receptors, Interleukin-2/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Diphtheria Toxin/administration & dosage , Female , Humans , Infusions, Intravenous , Interleukin-2/administration & dosage , Ligands , Lymphoma/metabolism , Male , Middle Aged , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , Treatment Outcome
16.
Cancer Biother Radiopharm ; 12(2): 79-87, 1997 Apr.
Article in English | MEDLINE | ID: mdl-10851451

ABSTRACT

Analyses were performed on 40 patients with TAG-72 expressing metastatic cancer who were entered into three phase II clinical trials. The dose selected was the maximum tolerated dose in phase I studies. Patients all had unresectable metastatic colon or prostate cancer and had recovered from prior therapies. Patients in trials #1 and #2 received 75 mCi/m2 131I-CC49 antibody whereas those in trial #3 received a total of 75 mCi/m2 with equal amounts of 131I-CC49 and 131I-COL-1. The three trials have resulted in a reproducible degree of reversible marrow suppression; 72.5% of patients experienced moderate or severe toxicity. Comparisons were made between demographic, clinical and pharmacokinetical variables and the grade of WBC toxicity, platelet toxicity and the sum of the two as total toxicity. Whole body radiation dose had a statistically significant relationship with platelet toxicity (r = 0.38, p = 0.015) and total toxicity (r = 0.34, p = 0.035). The bone marrow radiation dose is significantly related to all toxicity indicators with correlation coefficients with WBC and platelet toxicities of 0.47 (p = 0.002) and 0.34 (p = 0.033), respectively. Plasma half-life had the strongest correlation with WBC toxicity and combined toxicities. Multivariate models were developed to help describe the simultaneous effect of these variables on toxicity. The results show that the MTD dose was safely given to patients who varied in age, disease burden and degree of marrow compromise. This supports the contention that a fixed dose of radiolabeled antibody per body mass or m2 can be given to a diverse group of non-lymphoma patients with a predictable toxicity range.


Subject(s)
Antigens, Neoplasm/immunology , Colonic Neoplasms/radiotherapy , Glycoproteins/immunology , Prostatic Neoplasms/radiotherapy , Radioimmunotherapy/adverse effects , Adult , Aged , Female , Humans , Interleukin-1/adverse effects , Leukocytes/drug effects , Male , Middle Aged
17.
Neuroendocrinology ; 64(5): 364-78, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8930937

ABSTRACT

Based on previous findings that the inhibitory hypophysiotropic factor somatostatin (somatotropin-release-inhibiting hormone, SRIH) is markedly reduced in growth hormone (GH)-deficient transgenic or spontaneous Snell dwarf mice, the present study was undertaken to determine whether hypophysiotropic SRIH expression was reduced in a type of dwarf mouse (Ames, df/df) in which SRIH had not been assessed, and whether the supposed reduction was present throughout life or was the result of regression after initial normal differentiation. Brain sections from normal (DF/?) and df/df mice were immunostained for SRIH using both standard and 'Elite' avidin-biotin complex reagents (Vectastain kits, Vector Laboratories, Inc., Burlingame, Calif., USA). Selected adult mice were treated with intracerebroventricular colchicine to maximize SRIH retention in perikarya. The developmental pattern of hypophysiotropic SRIH was assessed in brains of DF/? and df/df mice at 1, 3, 7, 14, 21, 60, and 90 days (d) postnatally. SRIH-immunoreactive neurons in the anterior periventricular nucleus (PeN) were quantified at each age. Although the use of Elite reagents or Elite and colchicine pretreatment increased (p < 0.001) the number of immunoreactive cells that were detectable in adult (60- to 90-day-old) df/df mice, the number of PeN SRIH neurons was reduced to 28% (p < 0.01) in untreated, and to 47% (p < 0.01) in colchicine-treated, df/df compared with DF/?, mice. In other CNS areas, SRIH immunostaining was comparable for df/df and DF/? mice, including neuron numbers in the medial basal hypothalamus of untreated mice. In postnatal development, SRIH was detectable in median eminence (ME) terminals at birth in some mice of both phenotypes, and at 3 d in all DF/? mice; ME SRIH was detectable in all mice by 7 d. In PeN, SRIH cells were first detectable consistently in normals at 3 d, and in dwarfs at 7 d. In DF/? mice, numbers of immunoreactive SRIH perikarya increased from 3 to 21 d, then plateaued. In dwarfs, SRIH cell numbers increased through 14 d. Numbers of SRIH perikarya were lower in df/df than in DF/? at 7, 14, 21, 60, and 90 d (all p < 0.05 or less). Thus, in Ames dwarf mice, as in other GH-deficient models, SRIH is markedly reduced in hypophysiotropic, ME-projecting neurons. The developmental pattern of hypophysiotropic SRIH in Ames dwarf mice is different from that of hypophysiotropic dopaminergic (DA) neurons in these animals, which are also prolactin (PRL)-deficient. Although DA levels and cell numbers are reduced markedly in adult df/df mice, both parameters have been found to be comparable to those of DF/? mice for the first 2-3 weeks postnatally. The consistent PeN SRIH deficit in dwarfs may reflect the importance of GH feedback earlier in development, because GH production in normal mice begins before birth, whereas PRL is not detectable until 7 d postnatally. The findings indicate that absent GH production has a marked negative effect on differentiation and levels of peptide expression in hypophysiotropic SRIH neurons.


Subject(s)
Brain/growth & development , Dwarfism/metabolism , Growth Hormone/deficiency , Somatostatin/metabolism , Aging , Animals , Brain/cytology , Brain/drug effects , Brain Chemistry , Colchicine/pharmacology , Dwarfism/etiology , Indicators and Reagents , Median Eminence/chemistry , Mice , Mice, Mutant Strains , Neurons/chemistry , Paraventricular Hypothalamic Nucleus/chemistry , Somatostatin/analysis
18.
J Immunother Emphasis Tumor Immunol ; 18(4): 231-41, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8680651

ABSTRACT

In a phase I trial, eight patients with non-Hodgkin's B-cell lymphoma received mouse IgG1k monoclonal antibody HD37 specific for CD19 conjugated to deglycosylated ricin A chain (dgA) administered in four doses at 4-h intervals with total doses ranging from 4-12 mg/m2. This schedule generated serum levels of immunotoxin which were sustained over 36 h. The plasma half-life of HD37-dgA was 17 +/- 4 (SD) h. The HD37-dgA conjugate was stable in vivo as demonstrated by serum levels of HD37-dgA conjugate comparable to those of total HD37 antibody. Peak serum levels attained after the fourth dose ranged from 0.36 to 5.63 micrograms/ml. Two of seven evaluable patients developed modest human anti-immunotoxin antibody responses. Toxicity in patients 1-7 consisted of dose-dependent capillary leak syndrome with hypoalbuminemia, orthostatic hypotension, and weight gain. Patient 8 died on day 8 with severe capillary leak, bronchopneumonia, and rhabdomyolysis. All patients had progressive disease at 4 weeks except patient 8, who exhibited a near-complete remission before his death. This intensive schedule appears to produce inordinate toxicity with a maximal tolerated total dose of 8 mg/m2.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD19/immunology , Immunotoxins/administration & dosage , Immunotoxins/therapeutic use , Lymphoma, B-Cell/drug therapy , Ricin/therapeutic use , Adult , Aged , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Drug Administration Schedule , Female , Glycosylation , Humans , Immunotoxins/pharmacokinetics , Male , Middle Aged , Ricin/administration & dosage , Ricin/pharmacokinetics
19.
Cancer Res ; 55(19): 4339-46, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7671245

ABSTRACT

In a Phase II study, 14 patients with metastatic gastrointestinal cancer received the mAb D612 (40 mg/m2, days 4, 7, and 11) in combination with recombinant human monocyte colony-stimulating factor [(rhM-CSF) 80 micrograms/kg/days 1-14]. The combined treatment was well tolerated and resulted in characteristic biological activity associated with each of the agents. Thus, 10 of 14 patients experienced D612-associated secretory diarrhea, which responded to the prostaglandin inhibitor Indomethacin in 5 of 7 patients. rhM-CSF therapy was associated with peripheral monocytosis (peak absolute monocyte count, 1444 +/- 394/mm3) and thrombocytopenia (nadir count, 78 +/- 10/mm3). Monocyte surface marker analysis revealed a high baseline expression of CD16+ cells in our patient population with an additional increase with rhM-CSF therapy. We observed a correlation between the degree of thrombocytopenia and the pretreatment CD16+ monocyte count. Of the plasma cytokines assayed, serum Neopterin demonstrated the most consistent increase during rhM-CSF therapy. There was a significant difference in the half-life of the first and last dose of D612 (35.8 +/- 2 versus 27 +/- 2.9 h; P < 0.05). Eleven of fourteen patients developed low-moderate levels of anti-D612 antibody. Despite the observed biological activity of both rhM-CSF and D612 and the previously described in vitro synergy, no clinical antitumor responses were observed in this Phase II study.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Gastrointestinal Neoplasms/therapy , Macrophage Colony-Stimulating Factor/administration & dosage , Adult , Aged , Animals , Antibodies, Anti-Idiotypic/blood , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Blood Cell Count , Cytokines/blood , Female , Humans , Macrophage Colony-Stimulating Factor/adverse effects , Male , Mice , Middle Aged , Neoplasm Metastasis , Receptors, IgG/analysis , Recombinant Proteins/administration & dosage
20.
Blood ; 85(10): 2910-7, 1995 May 15.
Article in English | MEDLINE | ID: mdl-7742551

ABSTRACT

The small subset of circulating monocytes that express the maturation-associated CD16 antigen has recently been reported to be elevated in patients with bacterial sepsis. We now show that this novel CD16+ monocyte population is also spontaneously expanded in patients with cancer. We studied 14 patients with metastatic gastrointestinal carcinoma enrolled ina clinical trial of recombinant human macrophage colony-stimulating factor (rhMCSF) plus monoclonal antibody D612. We found that before any cytokine treatment, 12 of 14 patients constitutively displayed significant elevations in both the percentage and the absolute number of CD16+ monocytes, as compared with both normal subjects and ill patients with elevated monocyte counts but without malignancy. CD16+ monocytes accounted for 46% +/- 22% of total monocytes in the patients with cancer versus 5% +/- 3% for controls (P < .01). The increase was not attributable to infection or intercurrent illness and appeared to be associated with the underlying malignancy itself. A similar spontaneous elevation of CD16+ monocytes was observed in 35 of 44 additional patients diagnosed with a variety of other solid tumors. When patients with gastrointestinal carcinoma were treated with rhMCSF, there was a marked further increase in the percentage of CD16+ monocytes (to 83% +/- 11%), as well as in the absolute number of CD16+ cells and the level of CD16 antigen expression. In every case, the patients with cancer showed a greater CD16+ monocyte response than the maximal response obtained in normal volunteer subjects treated witha similar regimen of rhMCSF (n = 5, P < .001), suggesting that the presence of malignancy primed patients for enhanced responsiveness to rhMCSF. We hypothesize that spontaneous expansion of the CD16+ monocyte population may represent a novel biologic marker for a widespread and previously unsuspected host immune response to malignancy.


Subject(s)
Macrophage Colony-Stimulating Factor/pharmacology , Monocytes/cytology , Neoplasms/immunology , Receptors, IgG/metabolism , Adult , Flow Cytometry , Humans , Immunophenotyping , Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasm Metastasis , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL