scispace - formally typeset
Search or ask a question

Showing papers by "Pramod K. Srivastava published in 2003"


Journal ArticleDOI
TL;DR: This unit contains a table that lists common HSPs and summarizes their characteristics including (a) name, (b) subcellular localization, (c) known function, (d) chromosome assignment, (e) brief comments, and (f) references.
Abstract: Heat-shock proteins (HSPs), or stress proteins, are highly conserved and present in all organisms and in all cells of all organisms. Selected HSPs, also known as chaperones, play crucial roles in folding/unfolding of proteins, assembly of multiprotein complexes, transport/sorting of proteins into correct subcellular compartments, cell-cycle control and signaling, and protection of cells against stress/apoptosis. More recently, HSPs have been implicated in antigen presentation with the role of chaperoning and transferring antigenic peptides to the class I and class II molecules of the major histocompatibility complexes. In addition, extracellular HSPs can stimulate professional antigen-presenting cells of the immune system, such as macrophages and dendritic cells. HSPs constitute a large family of proteins that are often classified based on their molecular weight: hsp10, hsp40, hsp60, hsp70, hsp90, etc. This unit contains a table that lists common HSPs and summarizes their characteristics including (a) name, (b) subcellular localization, (c) known function, (d) chromosome assignment, (e) brief comments, and (f) references.

859 citations


Journal Article
TL;DR: In this paper, the authors tested in humans whether vaccination with HSPPC-gp96 (Oncophage) from autologous liver metastases of colorectal carcinoma induces cancer-specific T-cell responses in patients rendered disease free by surgery.
Abstract: Purpose: Heat shock proteins (HSP) from tumor cells contain the gp96 polypeptide associated with cancer-specific antigenic peptides. Mice that are immunized with HSP/peptide-complex (HSPPC) derived from cancer tissue reject tumor from which HSPs are purified. We tested in humans whether vaccination with HSPPC-gp96 (Oncophage) from autologous liver metastases of colorectal carcinoma induces cancer-specific T-cell responses in patients rendered disease free by surgery. Experimental Design: Twenty-nine consecutive patients underwent radical resection of liver metastases [Memorial Sloan-Kettering Cancer Center (MSKCC) score 1–3 (good prognosis), 18 patients; score 4–5 (bad prognosis), 11 patients] and received autologous tumor-derived HSPPC-96. Two vaccine cycles were administered (four weekly injections followed by four biweekly injections after 8 weeks). Class-I HLA-restricted, anti-colon cancer lines T-cell response was measured by ELISPOT assay on peripheral blood mononuclear cells (PBMCs) obtained before and after vaccination. Feasibility, safety, and possible clinical benefits were also evaluated. Results: Either a de novo induced or a significant increase of preexisting class I HLA-restricted T-cell-mediated anti-colon cancer response was observed in 15 (52%) of 29 patients. Frequency of CD3+, CD45RA+, and CCR7− T lymphocytes increased in immune responders. No relevant toxicity was observed. As expected, patients with good prognosis had a significantly better clinical outcome than those with poor prognosis [2-year overall survival (OS), 89 versus 64%, P = 0.001; disease-free survival (DFS), 46 versus 18%, P = 0.001]. Patients with immune response had a statistically significant clinical advantage over nonresponding subjects (2-year OS, 100% versus 50%, P = 0.001; DFS, 51% versus 8%, P = 0.0001). Occurrence of immune response led to better tumor-free survival, whatever the predicted prognosis was (hazard ratio, 0.11–0.12 with/without stratification; P = 0.0012–0.0003). Conclusions: HSPPC-96 vaccination after resection of colorectal liver metastases is safe and elicits a significant increase in CD8+ T-cell response against colon cancer. In this limited number of patients, two-year OS and DFS were significantly improved in subjects with postvaccination antitumor immune response, independently from other clinical prognostic factors.

251 citations


Journal ArticleDOI
TL;DR: Evidence is provided that hsp96 derived from human tumors can present antigenic peptides to CD8+ T cells and activate them both in vitro and in vivo, thus representing an important tool for vaccination in cancer patients.
Abstract: Heat shock proteins (hsp) 96 play an essential role in protein metabolism and exert stimulatory activities on innate and adaptive immunity. Vaccination with tumor-derived hsp96 induces CD8(+) T cell-mediated tumor regressions in different animal models. In this study, we show that hsp96 purified from human melanoma or colon carcinoma activate tumor- and Ag-specific T cells in vitro and expand them in vivo. HLA-A*0201-restricted CD8(+) T cells recognizing Ags expressed in human melanoma (melanoma Ag recognized by T cell-1 (MART-1)/melanoma Ag A (Melan-A)) or colon carcinoma (carcinoembryonic Ag (CEA)/epithelial cell adhesion molecule (EpCAM)) were triggered to release IFN-gamma and to mediate cytotoxic activity by HLA-A*0201-matched APCs pulsed with hsp96 purified from tumor cells expressing the relevant Ag. Such activation occurred in class I HLA-restricted fashion and appeared to be significantly higher than that achieved by direct peptide loading. Immunization with autologous tumor-derived hsp96 induced a significant increase in the recognition of MART-1/Melan-A(27-35) in three of five HLA-A*0201 melanoma patients, and of CEA(571-579) and EpCAM(263-271) in two of five HLA-A*0201 colon carcinoma patients, respectively, as detected by ELISPOT and HLA/tetramer staining. These increments in Ag-specific T cell responses were associated with a favorable disease course after hsp96 vaccination. Altogether, these data provide evidence that hsp96 derived from human tumors can present antigenic peptides to CD8(+) T cells and activate them both in vitro and in vivo, thus representing an important tool for vaccination in cancer patients.

123 citations


Journal ArticleDOI
TL;DR: The sequence of the Ecgp gene shows that it is highly similar to gp96, a tumor rejection antigen-1, and contains an endoplasmic reticulum retention signal, KDEL, and Northern blot analysis of total RNA from various eukaryotic cells indicates that Ecgp is significantly expressed in HBMECs.
Abstract: Escherichia coli is one of the most common gram-negative bacteria that cause meningitis in neonates. Our previous studies have shown that outer membrane protein A (OmpA) of E. coli interacts with a 95-kDa human brain microvascular endothelial cell (HBMEC) glycoprotein, Ecgp, for invasion. Here, we report the identification of a gene that encodes Ecgp by screening of an HBMEC cDNA expression library as well as by 5′ rapid amplification of cDNA ends. The sequence of the Ecgp gene shows that it is highly similar to gp96, a tumor rejection antigen-1, and contains an endoplasmic reticulum retention signal, KDEL. Overexpression of either Ecgp or gp96 in both HBMECs and CHO cells increases E. coli binding and invasion. We further show that Ecgp gene-transfected HBMECs express Ecgp on the cell surface despite the presence of the KDEL motif. Northern blot analysis of total RNA from various eukaryotic cells indicates that Ecgp is significantly expressed in HBMECs. Recombinant His-tagged Ecgp blocked E. coli invasion efficiently by binding directly to the bacteria. These results suggest that OmpA of E. coli K1 interacts with a gp96-like molecule on HBMECs for invasion.

88 citations


Journal ArticleDOI
TL;DR: Surprisingly, even brief exposure to elevated temperatures has a powerful effect on the immunostimulatory capacity of dendritic cells, a key antigen-presenting cell in the immune system.
Abstract: Fever is a phylogenetically conserved biological phenomenon and a common consequence of infection. Here, we examine in vitro and in vivo the effect of febrile temperature on dendritic cells (DC), a key antigen-presenting cell in the immune system. Elevated temperatures are observed to cause immature DC to mature, specifically through elevation of intracellular levels of hsp90. Surprisingly, even brief exposure to elevated temperatures has a powerful effect on the immunostimulatory capacity of DC. These results bear on the mechanisms of the salutary effects of fever as well as of behavioral elevations of temperature such as saunas and warm blankets.

84 citations


Journal ArticleDOI
15 May 2003-Blood
TL;DR: The ability of CD91 to internalize alpha-defensins and to cross-present exogenous antigen to cytotoxic T lymphocytes through major histocompatibility complex (MHC) class 1 may maintain CD8(+) responses in these patients.

56 citations


Journal ArticleDOI
TL;DR: The effects of PbO on optical properties of dysprosium doped calibo glass have been studied and the I-R absorption spectra of these glasses have been recorded and interpreted in terms of changes in the structure of the glasses.

54 citations


Journal ArticleDOI
01 Sep 2003-Blood
TL;DR: Despite the impaired functional capacity of dendritic cells (DCs) derived from patients with KS, DCs retain the ability to prime the adaptive arm of the immune system through the common HSP receptor, leading to phenotypic activation and stimulation of tetramer-positive CD8+ cytotoxic T cells.

44 citations


Journal Article
TL;DR: The number of ways in which cells die is broadly characterized into apoptosis and necrosis (see 1 for a more nuanced and accurate view).
Abstract: The number of ways in which cells die is broadly characterized into apoptosis and necrosis (see 1 for a more nuanced and accurate view). Apoptosis refers to programmed cell death, where exposure of a cell to events such as radiation, certain cytokines, or drugs leads to activation of a specific cascade of events that end in DNA fragmentation and a ‘clean’ death. The contents of the cell are not released into the external milieu but get packaged into apoptotic bodies which may be scavenged by neighboring phagocytes. End of story. Necrotic death on the other hand may result from unplanned events, such as lytic infection by a virus, loss of membrane permeability, mechanical tearing of a cell, etc. Such cell death leads to spilling of the cellular contents into the immediate milieu and is thus ‘not clean’.

43 citations


Patent
25 Apr 2003
TL;DR: In this article, a method of using heat shock proteins (HSPs) to amplify the immune response initiated by a vaccine is presented. But the method is not suitable for the treatment and prevention of cancer and infectious diseases in a subject.
Abstract: The present invention provides for a method of using heat shock proteins (HSPs) to amplify the immune response initiated by a vaccine. HSPs can be introduced into a subject before, concurrently, or after the administration of a vaccine. The HSPs can also be used to activate antigen presenting cells which are then introduced into a subject in conjunction with a vaccine. The HSPs used in the methods of the invention can be unbound or can be covalently or noncovalently bound to a peptide that is unrelated to the vaccine. The subject is preferably mammalian, and most preferably human. It is shown by way of example herein that HSPs induces secretion of cytokines and surface expression of antigen-presenting and co-stimulatory molecules. The invention also encompasses methods of treatment and prevention of cancer and infectious diseases in a subject.

37 citations


Patent
25 Apr 2003
TL;DR: In this paper, a heat shock protein (HSP) or α-2-macroglobulin (α2M) preparation is administered in conjunction with a non-vaccine treatment modality for the treatment of cancer or infectious diseases.
Abstract: The present invention relates to methods of improving a treatment outcome comprising administering a heat shock protein (HSP) preparation or an α-2-macroglobulin (α2M) preparation with a non-vaccine treatment modality. In particular, an HSP preparation or an α2M preparation is administered in conjunction with a non-vaccine treatment modality for the treatment of cancer or infectious diseases. In the practice of the invention, a preparation comprising HSPs such as but not limited to, hsp70, hsp90 and gp96 alone or in combination with each other, noncovalently or covalently bound to antigenic molecules or α2M, noncovalently or covalently bound to antigenic molecules is administered in conjunction with a non-vaccine treatment modality.

Patent
02 May 2003
TL;DR: In this paper, the authors proposed methods and pharmaceutical compositions useful for the prevention and treatment of any disease wherein the treatment of such disease would be improved by an enhanced immune response, such as infectious diseases, primary and metastatic neoplastic diseases (i.e., cancer), or neurodegenerative or amyloid diseases.
Abstract: The present invention relates to methods and pharmaceutical compositions useful for the prevention and treatment of any disease wherein the treatment of such disease would be improved by an enhanced immune response, such as infectious diseases, primary and metastatic neoplastic diseases (i.e., cancer), or neurodegenerative or amyloid diseases. In particular, the contemplated invention is directed to method comprising the administration of heat shock/stress proteins (HSPs) or HSP complexes alone or in combination with each other, in combination with the administration of an immunoreactive reagent. The invention also provides pharmaceutical compositions comprising one or more HSPs or HSP complexes in combination with an immunoreactive reagent. Additionally, the invention contemplates the use of the methods and compositions of the invention to enhance or improve passive immunotherapy and effector cell function.

01 Jan 2003
TL;DR: HSPPC-96 vaccination after resection of colorectal liver metastases is safe and elicits a significant increase in CD8+ T-cell response against colon cancer, and two-year OS and DFS were significantly improved in subjects with postvaccination antitumor immune response.
Abstract: Purpose: Heat shock proteins (HSP) from tumor cells contain the gp96 polypeptide associated with cancer-specific antigenic peptides. Mice that are immunized with HSP/peptide-complex (HSPPC) derived from cancer tissue reject tumor from which HSPs are purified. We tested in humans whether vaccination with HSPPC-gp96 (Oncophage) from autologous liver metastases of colorectal carcinoma induces cancer-specific T-cell responses in patients rendered disease free by surgery. Experimental Design: Twenty-nine consecutive patients underwent radical resection of liver metastases [Memorial Sloan-Kettering Cancer Center (MSKCC) score 1–3 (good prognosis), 18 patients; score 4–5 (bad prognosis), 11 patients] and received autologous tumor-derived HSPPC-96. Two vaccine cycles were administered (four weekly injections followed by four biweekly injections after 8 weeks). Class-I HLA-restricted, anti-colon cancer lines T-cell response was measured by ELISPOT assay on peripheral blood mononuclear cells (PBMCs) obtained before and after vaccination. Feasibility, safety, and possible clinical benefits were also evaluated. Results: Either a de novo induced or a significant increase of preexisting class I HLA-restricted T-cell-mediated anti-colon cancer response was observed in 15 (52%) of 29 patients. Frequency of CD3+, CD45RA+, and CCR7− T lymphocytes increased in immune responders. No relevant toxicity was observed. As expected, patients with good prognosis had a significantly better clinical outcome than those with poor prognosis [2-year overall survival (OS), 89 versus 64%, P = 0.001; disease-free survival (DFS), 46 versus 18%, P = 0.001]. Patients with immune response had a statistically significant clinical advantage over nonresponding subjects (2-year OS, 100% versus 50%, P = 0.001; DFS, 51% versus 8%, P = 0.0001). Occurrence of immune response led to better tumor-free survival, whatever the predicted prognosis was (hazard ratio, 0.11–0.12 with/without stratification; P = 0.0012–0.0003). Conclusions: HSPPC-96 vaccination after resection of colorectal liver metastases is safe and elicits a significant increase in CD8+ T-cell response against colon cancer. In this limited number of patients, two-year OS and DFS were significantly improved in subjects with postvaccination antitumor immune response, independently from other clinical prognostic factors.

Patent
28 Feb 2003
TL;DR: In this article, the authors provide methods for determining the biological activity of heat shock proteins or heat shock protein-peptide complexes based on the ATPase activity or the multimeric structure of the heat-shaking proteins.
Abstract: In one aspect, the invention provides methods for determining the biological activity of heat shock proteins or heat shock protein-peptide complexes based on the ATPase activity or the multimeric structure of the heat shock proteins or heat shock protein-peptide complexes, and methods for screening agents that modulate the biological activity of heat shock proteins or heat shock protein-peptide complexes. In another aspect, the invention provides complexes, compositions and methods for enhancing the immunogenicity of a heat shock protein or a complex comprising a heat shock protein and an antigenic molecule.

Patent
27 Oct 2003
TL;DR: In this article, the authors described a method for extracting a tissue sample from a patient or a tumor into a chamber of a treatment device and then lysing the tissue sample into an laysed tissue sample within the chamber of the treatment device.
Abstract: The treatment device includes an extraction mechanism, configured to extract a tissue sample from a patient or a tumor, coupled to a lysis mechanism. The lysis mechanism is configured to induce lysis of the tissue sample into a lysed tissue sample. The treatment device also includes an administration mechanism that is coupled to both the extraction mechanism and the lysis mechanism. Further included with the treatment device is an additive mechanism configured to add an additive solution to the lysed tissue sample before the lysed tissue sample is administered to the patient. Furthermore, the invention also provided a method for treating a cancer. The method includes the steps of extracting a tissue sample from a patient or a tumor into a chamber of a treatment device. Lysing the tissue sample into an laysed tissue sample within the chamber of the treatment device and administering the lysed tissue sample directly from the treatment device into the patient.

Patent
07 Oct 2003
TL;DR: In this paper, the use of natural and recombinant p95 forms and fragments as heat shock protein binding proteins is discussed, based on the discovery that a p95 can be recombinantly expressed.
Abstract: The present invention relates to compositions and methods for the use of natural and recombinant p95 forms and fragments as heat shock protein binding proteins. The invention is based, in part, on the Applicant's discovery that a p95 can be recombinantly expressed. The present invention also relates to CD91 polypeptide fragments that comprise at least p95 and additional contiguous sequence from domain II, III, and IV of CD91. The present invention provides nucleic acid molecules encoding a CD91 polypeptide fragment or an analog, derivative or mimetic thereof, CD91 polypeptide fragments, or analogs, derivatives or mimetics thereof, vectors comprising a nucleic acid molecule encoding a CD91 polypeptide fragment, expression vectors comprising a nucleic acid molecule encoding a CD91 polypeptide fragment, eukaryotic and prokaryotic cells recombinantly expressing a CD91 polypeptide fragment, methods of identifying compounds that interact with a CD91 polypeptide fragment or the interaction of a CD91 polypeptide fragment and CD91 ligands, methods for modulating an immune response with the compositions and methods of the invention, and methods for treatment using the compositions and methods disclosed herein.

Patent
27 Oct 2003
TL;DR: In this article, the authors described a method for extracting a tissue sample from a patient or a tumor into a chamber of a treatment device and then lysing the tissue sample into an laysed tissue sample within the chamber of the treatment device.
Abstract: The treatment device includes an extraction mechanism, configured to extract a tissue sample from a patient or a tumor, coupled to a lysis mechanism. The lysis mechanism is configured to induce lysis of the tissue sample into a lysed tissue sample. The treatment device also includes an administration mechanism that is coupled to both the extraction mechanism and the lysis mechanism. Further included with the treatment device is an additive mechanism configured to add an additive solution to the lysed tissue sample before the lysed tissue sample is administered to the patient. Furthermore, the invention also provided a method for treating a cancer. The method includes the steps of extracting a tissue sample from a patient or a tumor into a chamber of a treatment device. Lysing the tissue sample into an laysed tissue sample within the chamber of the treatment device and administering the lysed tissue sample directly from the treatment device into the patient.

Book ChapterDOI
01 Jan 2003
TL;DR: The connection between HSPs and tumor rejection antigens became apparent through tumor rejection studies in mice and rats and immunization of mice with HSP preparations from normal tissues or from antigenically-distinct tumor cells did not protect the animals against tumor challenge.
Abstract: The connection between HSPs and tumor rejection antigens became apparent through tumor rejection studies in mice and rats. In search for individually distinct tumor rejection antigens, tumor cell lysates were fractionated biochemically and each fraction was tested for its ability to immunize in vivo against the tumor. The tumor rejection antigens thus found were mostly HSPs (Tab. 1). Preparations of HSPs, e.g., HSP70, HSP90, gp96 and calreticulin (CRT) from Meth A fibrosarcoma (tumor induced in BALB/c mice by methylcholanthrene) when used to immunize BALB/c mice, rendered the mice immune to subsequent challenge with live Meth A tumor cells in a classical tumor rejection assay [1-3]. However, immunization of mice with HSP preparations from normal tissues [2] or from antigenically-distinct tumor cells [1, 4] did not protect the animals against tumor challenge.