Network


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

Hotspot


Dive into the research topics where Piotr J. Wysocki is active.

Publication


Featured researches published by Piotr J. Wysocki.


Expert Review of Molecular Diagnostics | 2010

Obesity, hyperinsulinemia and breast cancer: novel targets and a novel role for metformin.

Piotr J. Wysocki; Bogna Wierusz-Wysocka

The relationship between obesity, metabolic syndrome, diabetes and cancer has been recognized for many years. Multiple studies conducted in the last 20 years have identified molecular mechanisms responsible for this phenomenon. Elucidation of the important role of insulin, IGF receptor, mTOR and AMP-activated protein kinase in breast cancer biology has led to the development and subsequent clinical evaluation of novel targeted therapies, including IGF-1 receptor-specific antibodies or tyrosine kinase inhibitors and inhibitors of mTOR. There is also a growing interest in the use of metformin, which has been shown to possess antitumor activity resulting from activation of AMP-activated protein kinase and subsequent inhibiton of mTOR, as well as from decreased circulating insulin levels. Metformin has been shown to inhibit proliferation, invasion and angiogenesis of neoplastic cells and to overcome resistance of breast cancer to chemotherapy, hormonal therapy and HER2 inhibition. Recently, metformin has been demonstrated to inhibit breast cancer stem cell growth and to synergize with chemotherapy in suppression of tumor growth and prolongation of survival of breast tumor-bearing animals. Several currently ongoing Phase II and III clinical studies are evaluating the therapeutic efficacy of metformin in the treatment of early and advanced breast cancer patients.


Clinical Cancer Research | 2004

CpG Immunostimulatory Oligodeoxynucleotide 1826 Enhances Antitumor Effect of Interleukin 12 Gene-Modified Tumor Vaccine in a Melanoma Model in Mice

Tomasz Switaj; Ahmad Jalili; Anna Jakubowska; Nadzieja Drela; Magdalena Stoksik; Dominika Nowis; Grzegorz W. Basak; Jakub Golab; Piotr J. Wysocki; Andrzej Mackiewicz; Agata Sasor; Koryna Socha; Marek Jakóbisiak; Witold Lasek

Purpose: The effectiveness of interleukin (IL)-12-secreting tumor vaccines in the treatment of mouse tumors could be enhanced by concurrent application of cytokines and costimulatory molecules. We investigated the therapeutic potential of IL-12 gene-transduced melanoma vaccine in combination with CpG immunostimulatory oligodeoxynucleotide (ODN) 1826, an adjuvant known to favor development of Th1-biased immune response, in a B78-H1 (B78) melanoma model in mice. Experimental Design: Mice injected with B78 melanoma cells were treated with irradiated IL-12 gene-transduced B78 cells [B78/IL-12(X)] and/or ODN 1826. Mechanisms responsible for the antitumor effects of the treatment were investigated using fluorescence-activated cell sorter analysis, a standard 51Cr releasing assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and ELISA. Results: Single injection of B78/IL-12(X) cells had no effect on tumor growth, whereas seven consecutive daily injections of ODN 1826 markedly inhibited tumor progression with occasional curative effects. When used in combination, B78/IL-12(X) cells and ODN 1826 caused additional tumor growth reduction and eradication of tumors in 62% of treated mice. The combined treatment activated local inflammatory response against tumor but also induced systemic antitumor immunity. In vitro studies have shown that when used together, B78/IL-12(X) cells and ODN 1826 induced a potent Th1 response and suggested the role of IFN-γ in activation of the host immune response. The antitumor effects in double-treated mice were accompanied by the development of cytotoxic effectors in the spleen and activation of macrophages. Conclusions: The results provided the evidence that the combination of IL-12 gene-modified melanoma vaccine and ODN 1826 induces synergistically systemic and local antitumor immunity.


Oncologist | 2012

Correlation Between Quantitative HER-2 Protein Expression and Risk for Brain Metastases in HER-2+ Advanced Breast Cancer Patients Receiving Trastuzumab-Containing Therapy

Renata Duchnowska; Wojciech Biernat; Barbara Szostakiewicz; Jeff Sperinde; Fanny Piette; Mojgan Haddad; Agnes Paquet; Yolanda Lie; Bogumiła Czartoryska-Arłukowicz; Piotr J. Wysocki; Tomasz Jankowski; Barbara Radecka; Małgorzata Foszczyńska-Kłoda; Maria Litwiniuk; Sylwia Dȩbska; Jodi Weidler; Weidong Huang; Marc Buyse; Michael Bates; Jacek Jassem

BACKGROUND Patients with human epidermal growth factor receptor (HER)-2+ breast cancer are at particularly high risk for brain metastases; however, the biological basis is not fully understood. Using a novel HER-2 assay, we investigated the correlation between quantitative HER-2 expression in primary breast cancers and the time to brain metastasis (TTBM) in HER-2+ advanced breast cancer patients treated with trastuzumab. METHODS The study group included 142 consecutive patients who were administered trastuzumab-based therapy for HER-2+ metastatic breast cancer. HER-2/neu gene copy number was quantified as the HER-2/centromeric probe for chromosome 17 (CEP17) ratio by central laboratory fluorescence in situ hybridization (FISH). HER-2 protein was quantified as total HER-2 protein expression (H2T) by the HERmark® assay (Monogram Biosciences, Inc., South San Francisco, CA) in formalin-fixed, paraffin-embedded tumor samples. HER-2 variables were correlated with clinical features and TTBM was measured from the initiation of trastuzumab-containing therapy. RESULTS A higher H2T level (continuous variable) was correlated with shorter TTBM, whereas HER-2 amplification by FISH and a continuous HER-2/CEP17 ratio were not predictive (p = .013, .28, and .25, respectively). In the subset of patients that was centrally determined by FISH to be HER-2+, an above-the-median H2T level was significantly associated with a shorter TTBM (hazard ratio, [HR], 2.4; p = .005), whereas this was not true for the median HER-2/CEP17 ratio by FISH (p = .4). Correlation between a continuous H2T level and TTBM was confirmed on multivariate analysis (HR, 3.3; p = .024). CONCLUSIONS These data reveal a strong relationship between the quantitative HER-2 protein expression level and the risk for brain relapse in HER-2+ advanced breast cancer patients. Consequently, quantitative assessment of HER-2 protein expression may inform and facilitate refinements in therapeutic treatment strategies for selected subpopulations of patients in this group.


Clinical Cancer Research | 2014

Quantitative Measurements of Tumoral p95HER2 Protein Expression in Metastatic Breast Cancer Patients Treated with Trastuzumab: Independent Validation of the p95HER2 Clinical Cutoff

Renata Duchnowska; Jeff Sperinde; Ahmed Chenna; Mojgan Haddad; Agnes Paquet; Yolanda Lie; Jodi Weidler; Weidong Huang; John Winslow; Tomasz Jankowski; Bogumiła Czartoryska-Arłukowicz; Piotr J. Wysocki; Małgorzata Foszczyńska-Kłoda; Barbara Radecka; Maria Litwiniuk; Jolanta Żok; Michał Wiśniewski; Dorota Zuziak; Wojciech Biernat; Jacek Jassem

Purpose: P95HER2 (p95) is a truncated form of the HER2, which lacks the trastuzumab-binding site and contains a hyperactive kinase domain. Previously, an optimal clinical cutoff of p95 expression for progression-free survival (PFS) and overall survival (OS) was defined using a quantitative VeraTag assay (Monogram Biosciences) in a training set of trastuzumab-treated metastatic breast cancer (MBC) patients. Experimental Design: In the current study, the predictive value of the p95 VeraTag assay cutoff established in the training set was retrospectively validated for PFS and OS in an independent series of 240 trastuzumab-treated MBC patients from multiple institutions. Results: In the subset of 190 tumors assessed as HER2-total (H2T)-positive using the quantitative HERmark assay (Monogram Biosciences), p95 VeraTag values above the predefined cutoff correlated with shorter PFS (HR = 1.43; P = 0.039) and shorter OS (HR = 1.94; P = 0.0055) where both outcomes were stratified by hormone receptor status and tumor grade. High p95 expression correlated with shorter PFS (HR = 2.41; P = 0.0003) and OS (HR = 2.57; P = 0.0025) in the hormone receptor-positive subgroup of patients (N = 78), but not in the hormone receptor-negative group. In contrast with the quantitative p95 VeraTag measurements, p95 immunohistochemical expression using the same antibody was not significantly correlated with outcomes. Conclusions: The consistency in the p95 VeraTag cutoff across different cohorts of patients with MBC treated with trastuzumab justifies additional studies using blinded analyses in larger series of patients. Clin Cancer Res; 20(10); 2805–13. ©2014 AACR.


Expert Opinion on Biological Therapy | 2004

Human cancer gene therapy with cytokine gene-modified cells

Piotr J. Wysocki; Aldona Karczewska-Dzionk; Małgorzata Mackiewicz-Wysocka; Andrzej Mackiewicz

Cytokines can impede tumour growth and activate innate and adaptive immune responses, leading to elimination of cancer cells. For many years, it was believed that systemic administration of recombinant cytokines might become a standard treatment of different cancer types. However, due to a high toxicity of therapeutic doses and a low efficacy, even in combination with chemotherapy, this strategy is generally not accepted. On the other hand, cancer gene therapy approaches utilising cells modified with cytokine genes seem to represent a novel promising approach. For the last decade, numerous Phase I and II clinical trials evaluating different therapies based on cytokine gene-modified cells have been carried out. In the early studies, several strategies have been shown to improve clinical outcomes and induce strong antitumour immune responses. Recently, a few prospective, randomised, Phase III clinical trials have been initiated in order to finally determine the efficacy of particular cancer immunogene therapy strategies. This article reviews the present status and perspectives of clinical trials of cancer immunotherapies utilising cytokine gene-modified cells.


Expert Opinion on Biological Therapy | 2001

Genetically modified tumour vaccines: an obstacle race to break host tolerance to cancer.

Sergiusz Nawrocki; Piotr J. Wysocki; Andrzej Mackiewicz

The development of genetically modified tumour vaccines (GMTV) has been prompted by a better understanding of antitumour immune responses and genetic engineering technologies, as well as the identification of numerous tumour antigens (TA) in several malignancies which occasionally induce spontaneous tumour regressions. Cellular vaccines are based on autologous or allogeneic tumour cells genetically engineered to secrete different cytokines, co-stimulatory molecules, or allogeneic HLA molecules in order to provide a strong stimulatory signal together with the presented TA. Another promising approach that is targeted towards breaking immune tolerance to TA, exploits dendritic cells (DC) loaded or genetically modified with TA (and sometimes cytokines). Effective nonviral and viral gene delivery systems have been constructed including a third generation of adenoviral, lentiviral and hybrid vectors. Studies in mice demonstrated that therapeutic, curative immune responses might be elicited by GMTV. Promising results from animal studies are rarely seen in human trials. Several reasons, such as numerous escape mechanisms of slowly evolving spontaneous tumours and immune incompetence of advanced patients, are major concerns. Improved monitoring of immune responses to GMTV is essential to distinguish between responders and non-responders in order to tailor immune therapy strategy to the individual patient.


Clinical Cancer Research | 2006

Captopril, an Angiotensin-Converting Enzyme Inhibitor, Promotes Growth of Immunogenic Tumors in Mice

Piotr J. Wysocki; Eliza Kwiatkowska; Urszula Kazimierczak; Wiktoria Suchorska; Dariusz W. Kowalczyk; Andrzej Mackiewicz

Purpose: Antitumor potential of angiotensin-converting enzyme inhibitors has been shown in different preclinical settings, which always involved immunocompromised organisms or nonimmunogenic tumor models. In our study, we wanted to evaluate the effect of captopril on growth of immunogenic tumors in immunocompetent animals. Experimental Design: We used different murine tumor models to evaluate the effect of captopril on tumor take and survival of tumor-bearing immunocompetent and immunocompromised mice. We used an orthotopic renal cell cancer model and highly immunogenic tumor model, which were based on kidney subcapsular injection of RenCa cells or s.c. injection of MethA cells, respectively. To show the influence of captopril on antigen-specific immune responses, we have used two model antigens (green fluorescent protein and β-galactosidase). Results: Captopril decreased survival of RenCa-bearing, immunocompetent mice in a dose-dependent manner and in adjuvant setting. In nephrectomized mice, captopril shortened their survival. Captopril promoted formation of immunogenic MethA sarcoma tumors but had no effect on nonimmunogenic melanoma cells (B78-H1). Treatment of immunocompromised mice bearing MethA tumors or RenCa kidney tumors with captopril did not affect tumor formation nor survival, respectively. Captopril-treated mice immunized with AdLacZ or AdGFP vectors did not generate or generated decreased numbers of antigen-specific CD8+ T cells, respectively. However, they showed B-cell responses represented by infiltration of MethA tumors with activated B cells and dramatically increased serum level of β-galactosidase-specific antibodies. Conclusions: Our results show a novel role of captopril in tumor biology and the tumor-promoting properties of captopril seem to be associated with its immunomodulatory potential.


Expert Opinion on Biological Therapy | 2002

Genetically modified dendritic cells - a new, promising cancer treatment strategy?

Piotr J. Wysocki; Piotr Grabarczyk; Małgorzata Mackiewicz-Wysocka; Dw Kowalczyk; Andrzej Mackiewicz

Dendritic cells (DCs), the most potent antigen-presenting cells (APCs), were discovered almost 30 years ago. Due to the priming of antigen-specific immune responses mediated by CD4+ and CD8+ lymphocytes, DCs are crucial for the induction of adaptive immunity against cancer. Therefore, vaccination of cancer patients with DCs presenting tumour-associated antigens (TAAs) have been believed to be a promising anticancer strategy. Multiple clinical trials have been carried out in order to evaluate the safety and efficacy of cancer vaccines based on antigen-pulsed DCs. However, pulsing of DCs with particular peptides has several disadvantages: i) short-time duration of antigen–major histocompatability complex (MHC) complexes, ii) a requirement for matching defined peptides with MHC complexes and iii) exclusive presentation of single antigen epitopes. Application of gene transfer technologies in the field of DC-based vaccines made possible the development of novel, anticancer immunisation strategies. In several animal models, DCs modified with genes encoding TAA or immunostimulatory proteins have been shown to be effective in the induction of antitumour immune responses. Based on these encouraging results, a first clinical trial of prostate cancer patients vaccinated with gene modified DCs has recently been initiated. In this article, methods used for genetic modification of DCs and anticancer vaccination strategies based on genetically modified DCs are reviewed.


Expert Opinion on Biological Therapy | 2007

Recent developments in renal cell cancer immunotherapy

Piotr J. Wysocki; J Zolnierek; C Szczylik; A Mackiewicz

Various immunotherapeutic approaches for the treatment of renal cell carcinoma (RCC) have been developed for > 90 years. Existing immunotherapeutic strategies against RCC include: systemic administration of cytokines; therapeutic vaccines based on tumor cells or dendritic cells; monoclonal antibodies; and adoptive immunotherapy (T cell transfer or allogeneic hematopoietic cell transplantation). However, the overall efficacy of immunotherapy for advanced RCC remains moderate. With the advent of molecularly targeted biological therapies that turned out to be significantly effective in the treatment of metastatic RCC, to many oncologists immunotherapy may seem to be moving into the periphery of RCC treatment strategies. However, for the last 2 years there has been significant progress made in immunotherapeutic approaches for the treatment of RCC. Immunotherapy still remains the only systemic therapeutic strategy that is believed to potentially cure RCC patients. The development of active and passive specific immunotherapeutic approaches, along with the possibility to ‘switch off’ particular immunosuppressive mechanisms (e.g., elimination of regulatory T cells, blockage of cytotoxic T lymphocyte antigen-4 signaling), have paved the way for future trials of new immunotherapies of RCC. However, the new studies will have to enroll optimally selected patients (nephrectomized, with non-massive metastases and good performance status) and will use tumor response criteria that are specifically optimized for clinical trials of immunotherapy.


Reports of Practical Oncology & Radiotherapy | 2002

Cancer gene therapy – state-of-the-art

Piotr J. Wysocki; Małgorzata Mackiewicz-Wysocka; Andrzej Mackiewicz

Abstract A number of gene therapy clinical trials are being carried out the world over. Gene therapy is being applied in (I) cancer diseases, involving the largest number of patients, (II) monogenic diseases, (III) infectious diseases, (IV) vascular diseases, (V) autoimmune diseases and others. In the last decade, several strategies of cancer gene therapy have emerged due to a rapid development of gene delivery systems, both viral (recombinant retroviruses, adenoviruses, AAVs, herpes viruses) and nonviral (liposomes, gene guns, electroporation). To date four main strategies of cancer gene therapy have been evaluated in clinical trials: (I) immunogene therapy, (II) suicide gene therapy, (III) antiangiogenic gene therapy, (IV) and administration of tumour suppressor genes. These strategies mostly involve: malignant melanoma, prostate cancer, renal cell cancer, colon cancer, breast and ovarian cancers, lung cancers, neoplastic diseases of the blood and brain tumours. At the Department of Cancer Immunology at the GreatPoland Cancer Center Gene Modified Tumour Vaccine has been tested in malignant melanoma patients for more than six years. Due to encouraging results from phase I and II of clinical trials a phase III was designed and will be started in 2003.

Collaboration


Dive into the Piotr J. Wysocki's collaboration.

Top Co-Authors

Avatar

Andrzej Mackiewicz

Poznan University of Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Piotr Potemski

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Maria Litwiniuk

Poznan University of Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Krzysztof Krzemieniecki

Jagiellonian University Medical College

View shared research outputs
Top Co-Authors

Avatar

Jacek Fijuth

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Lidia Rudnicka

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Arkadiusz Jeziorski

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomasz Świtaj

Medical University of Warsaw

View shared research outputs
Researchain Logo
Decentralizing Knowledge