Network


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

Hotspot


Dive into the research topics where John L. Herrmann is active.

Publication


Featured researches published by John L. Herrmann.


Clinical Cancer Research | 2006

CR011, a fully human monoclonal antibody-auristatin E conjugate, for the treatment of melanoma.

Kam Fai Tse; Michael Jeffers; Vincent A. Pollack; Denise A. McCabe; Melanie Shadish; Nikolai V. Khramtsov; Craig Hackett; Suresh Shenoy; Bing Kuang; Ferenc L. Boldog; John R. Macdougall; Luca Rastelli; John L. Herrmann; Michael A. Gallo; Gadi Gazit-Bornstein; Peter D. Senter; Damon L. Meyer; Henri Lichenstein; William J. LaRochelle

PURPOSE: Advanced melanoma is a highly drug-refractory neoplasm representing a significant unmet medical need. We sought to identify melanoma-associated cell surface molecules and to develop as well as preclinically test immunotherapeutic reagents designed to exploit such targets. EXPERIMENTAL DESIGN AND RESULTS: By transcript profiling, we identified glycoprotein NMB (GPNMB) as a gene that is expressed by most metastatic melanoma samples examined. GPNMB is predicted to be a transmembrane protein, thus making it a potential immunotherapeutic target in the treatment of this disease. A fully human monoclonal antibody, designated CR011, was generated to the extracellular domain of GPNMB and characterized for growth-inhibitory activity against melanoma. The CR011 monoclonal antibody showed surface staining of most melanoma cell lines by flow cytometry and reacted with a majority of metastatic melanoma specimens by immunohistochemistry. CR011 alone did not inhibit the growth of melanoma cells. However, when linked to the cytotoxic agent monomethylauristatin E (MMAE) to generate the CR011-vcMMAE antibody-drug conjugate, this reagent now potently and specifically inhibited the growth of GPNMB-positive melanoma cells in vitro. Ectopic overexpression and small interfering RNA transfection studies showed that GPNMB expression is both necessary and sufficient for sensitivity to low concentrations of CR011-vcMMAE. In a melanoma xenograft model, CR011-vcMMAE induced significant dose-proportional antitumor effects, including complete regressions, at doses as low as 1.25 mg/kg. CONCLUSION: These preclinical results support the continued evaluation of CR011-vcMMAE for the treatment of melanoma.


Oncogene | 1998

Prostate carcinoma cell death resulting from inhibition of proteasome activity is independent of functional Bcl-2 and P53

John L. Herrmann; Fermin Briones; Shawn Brisbay; Christopher J. Logothetis; Timothy J. McDonnell

The ATP/ubiquitin-dependent 26S proteasome is a central regulator of cell cycle progression and stress responses. While investigating the application of peptide aldehyde proteasome inhibitors to block signal-induced IκBα degradation in human LNCaP prostate carcinoma cells, we observed that persistent inhibition of proteasomal activity signals a potent cell death program. Biochemically, this program included substantial upregulation of PAR-4 (prostate apoptosis response-4), a putative pro-apoptotic effector protein and stabilization of c-jun protein, a potent pro-death effector in certain cells. We also observed modest downregulation of bcl-XL, a pro-survival effector protein. However, in contrast to some recent reports stable, high level, expression of functional bcl-2 protein in prostate carcinoma cells failed to signal protection against cell death induction by proteasome inhibitors. Also in disagreement to a recent report, no evidence was found for activation of the JNK stress kinase pathway. A role for p53, a protein regulated by the proteasome pathway, was ruled out, since comparable cell death induction by proteasome inhibitors occurred in PC-3 cells that do not express functional p53 protein. These data signify that the ubiquitin/proteasome pathway represents a potential therapeutic target for prostate cancers irrespective of bcl-2 expression or p53 mutations.


Clinical & Experimental Metastasis | 1995

The role of trophic factors and autocrine/paracrine growth factors in brain metastasis

David G. Menter; John L. Herrmann; Garth L. Nicolson

The brain is a unique microenvironment enclosed by the skull, lacking lymphatic drainage and maintaining i highly regulated vascular transport barrier. To metastasize to the brain malignant tumor cells must attach to microvessel endothelial cells, respond to brain-derived invasion factors, invade the blood-brain barrier and respond to survival and growth factors. Trophic factors are important in brain invasion because they can act to stimulate this process. In responsive malignant cells trophic factors such as neurotrophins can promote invasion by enhancing the production of basement membrane-degradative enzymes (such as type IV collagenase/gelatinase and heparanase) capable of locally destroying the basement membrane and the blood-brain barrier. We examined human melanoma cell lines that exhibit varying abilities to form brain metastases. These melanoma lines express low-affinity neurotrophin receptor p75NTR in relation to their brain-metastatic potentials but the variants do not express trkA, the gene encoding a high affinity nerve growth factor (NGF) tyrosine kinase receptor p140trkA. Melanoma cells metastatic to brain also respond to paracrine factors made by brain cells. We have found that a paracrine form of transferrin is important in brain metastasis, and brain-metastatic cells respond to low levels of transferrin and express high levels of transferrin receptors. Brain-metastatic tumor cells can also produce autocrine factors any inhibitors that influence their growth, invasion and survival in the brain. We found that brain-metastatic melanoma cells synthesize transcripts for the following autocrine growth factors: TGFβ, bFGF, TGFα and IL-1β. Synthesis of these factors may influence the production of neurotrophins by adjacent brain cells, such as oligodendrocytes and astrocytes. Increased amounts of NGF were found in tumor-adjacent tissues at the invasion front of human melanoma tumors in brain biopsies. Trophic factors, autocrine growth factors, paracrine growth factors and other factors may determine whether metastatic cells can successfully invade, colonize and grow in the central nervous system.


Current Topics in Microbiology and Immunology | 1996

Brain Metastasis: Role of Trophic, Autocrine, and Paracrine Factors in Tumor Invasion and Colonization of the Central Nervous System

Garth L. Nicolson; David G. Menter; John L. Herrmann; Zhong Yun; Philip G. Cavanaugh; Dario Marchetti

The brain is a unique target for tumor invasion and metastasis formation (Steck and Nicolson 1993; Nicolson 1993a; Menter et al. 1995a; Nicolson et al. 1994b. The central nervous system (CNS) is confined by the skull, and the brain is highly sensitive to the slightest change in the local microenvironment. The brain is also surrounded by a formidable blood-brain barrier (BBB), which does not allow penetration of most cell types. Because the brain lacks lymphatic drainage, cerebral edema is a major complication resulting from tumors in the CNS. When tumors form in the brain, either from endogenous brain cells or from metastases invading the BBB, they are very difficult to successfully treat. Therapy of brain tumors and metastases are often only palliative and are often accompanied by additional complications. A relatively small tumor in the brain cavity can cause severe symptoms, including impaired cognition, headaches, seizures, and eventually paralysis.


Journal of Neuro-oncology | 1994

Malignant melanoma metastasis to brain: role of degradative enzymes and responses to paracrine growth factors

Garth L. Nicolson; Motowo Nakajima; John L. Herrmann; David G. Menter; Philip G. Cavanaugh; June Sik Park; Dario Marchetti

Mouse and human melanoma cells metastatic to the brain express degradative enzyme activities that are used for invasion of brain basement membrane and parenchyma. Compared to poorly metastatic or lung- or ovary-metastatic murine melanoma lines, the brain-metastatic sublines secreted higher levels of a variety of degradative enzymes. Brain-metastatic murine and human melanoma cells also degraded subendothelial basement membrane and reconstituted basement membrane at rates higher than other metastatic melanoma cells. In some cases these degradative activities in mouse and human melanoma cells can be induced by paracrine factors known to be present in the brain parenchyma, such as nerve growth factor (NGF). NGF stimulates the expression of degradative enzymes, such as the endo-Β-glucuronidase heparanase, that are important in basement membrane penetration but this factor does not stimulate melanoma cell growth. The growth of brain-metastasizing melanoma cells appears to be stimulated by other paracrine growth factors, such as paracrine transferrin. Melanoma cells metastatic to brain express higher numbers of transferrin receptors and respond and proliferate at lower concentrations of transferrin than do melanoma cells metastatic to other sites or poorly metastatic melanoma cells. The results suggest that degradation and invasion of brain basement membrane and responses to paracrine neurotrophins and paracrine transferrins are important properties in brain metastasis of murine and human malignant melanoma cells.


Archive | 2002

Proteins and nucleic acids encoding same

Luca Rastelli; John L. Herrmann; John R. Macdougall; Haihong Zhong


Molecular Biology of the Cell | 1993

Mediation of NGF-stimulated extracellular matrix invasion by the human melanoma low-affinity p75 neurotrophin receptor: melanoma p75 functions independently of trkA.

John L. Herrmann; David G. Menter; Jun-ichi Hamada; Dario Marchetti; Motowo Nakajima; Garth L. Nicolson


Experimental Cell Research | 1997

Bcl-2 suppresses apoptosis resulting from disruption of the NF-κB survival pathway

John L. Herrmann; Alexander Beham; Mona Sarkiss; Paul J. Chiao; M. Todd Rands; Elizabeth M. Bruckheimer; Shawn Brisbay; Timothy J. McDonnell


Clinical Cancer Research | 2000

E1A-mediated paclitaxel sensitization in HER-2/neu-overexpressing ovarian cancer SKOV3.ip1 through apoptosis involving the caspase-3 pathway.

Naoto Ueno; Chandra Bartholomeusz; John L. Herrmann; Zeev Estrov; Ruping Shao; Michael Andreeff; Janet E. Price; Ralph Paul; Pervin Anklesaria; Dihua Yu; Mien Chie Hung


Oncogene | 1997

Bcl-2 inhibits p53 nuclear import following DNA damage

Alexander Beham; Maria C. Marin; Antonio Fernandez; John L. Herrmann; Shawn Brisbay; Ana M. Tari; Gabriel Lopez-Berestein; Guillermina Lozano; Mona Sarkiss; Timothy J. McDonnell

Collaboration


Dive into the John L. Herrmann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ferenc L. Boldog

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Michael Jeffers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William J. LaRochelle

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Garth L. Nicolson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David G. Menter

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge