Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Target Oncol ; 18(3): 391-402, 2023 05.
Article in English | MEDLINE | ID: mdl-37060431

ABSTRACT

BACKGROUND: Phosphatase of regenerating liver-3 (PRL-3) is involved in cellular processes driving metastasis, cell proliferation, invasion, motility and survival. It has been shown to be upregulated and overexpressed in cancer tissue, in contrast to low or no expression in most normal tissue. PRL3-zumab is a first-in-class humanized antibody that specifically binds to PRL-3 oncotarget with high affinity and has been shown to reduce tumor growth and increase survival. OBJECTIVE: In the study, we aimed to determine the safety and efficacy of PRL3-zumab in patients with advanced solid tumors and hematological malignancies. METHODS: We conducted a phase I, first-in-human study in advanced solid tumors and hematological malignancies to investigate the safety, tolerability and efficacy of PRL3-zumab. Response rates were evaluated using the Response Evaluation Criteria in Solid Tumors (RECIST) guideline (version 1.1) for solid tumors. For acute myeloid leukemia (AML) patients, bone marrow response criteria based on the European Leukaemia Network (ELN) 2017 guidelines for AML were used. We also explored the pharmacokinetics and pharmacodynamic relationships of PRL3-zumab in patients. This study was registered with ClinicalTrials.gov: NCT03191682. RESULTS: In the dose-escalation cohort, 11 patients with advanced solid tumors were enrolled into the study. An additional 12 patients with solid tumors and four patients with AML were enrolled in the dose-expansion cohort. Maximum tolerability was not achieved in this study, as there were no dose-limiting toxicities. Potential treatment-emergent adverse events were grade 1 increased stoma output and fatigue and grade 2 vomiting. Best response observed was stable disease in three solid-tumor patients (11.1%). The pharmacokinetics of PRL3-zumab were dose proportional, consistent with an IgG type monoclonal antibody. CONCLUSIONS: PRL3-zumab, a first-in-class humanized antibody, was safe and tolerable in solid tumors and hematological malignancies.


Subject(s)
Antineoplastic Agents , Hematologic Neoplasms , Leukemia, Myeloid, Acute , Neoplasms , Humans , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Hematologic Neoplasms/drug therapy , Maximum Tolerated Dose
4.
Commun Biol ; 4(1): 923, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326464

ABSTRACT

PRL3, a unique oncotarget, is specifically overexpressed in 80.6% of cancers. In 2003, we reported that PRL3 promotes cell migration, invasion, and metastasis. Herein, firstly, we show that PRL3 induces Polyploid Giant Cancer Cells (PGCCs) formation. PGCCs constitute stem cell-like pools to facilitate cell survival, chemo-resistance, and tumor relapse. The correlations between PRL3 overexpression and PGCCs attributes raised possibilities that PRL3 could be involved in PGCCs formation. Secondly, we show that PRL3+ PGCCs co-express the embryonic stem cell markers SOX2 and OCT4 and arise mainly due to incomplete cytokinesis despite extensive DNA damage. Thirdly, we reveal that PRL3+ PGCCs tolerate prolonged chemotherapy-induced genotoxic stress via suppression of the pro-apoptotic ATM DNA damage-signaling pathway. Fourthly, we demonstrated PRL3-zumab, a First-in-Class humanized antibody drug against PRL3 oncotarget, could reduce tumor relapse in 'tumor removal' animal model. Finally, we confirmed that PGCCs were enriched in relapse tumors versus primary tumors. PRL3-zumab has been approved for Phase 2 clinical trials in Singapore, US, and China to block all solid tumors. This study further showed PRL3-zumab could potentially serve an 'Adjuvant Immunotherapy' after tumor removal surgery to eliminate PRL3+ PGCC stem-like cells, preventing metastasis and relapse.


Subject(s)
Giant Cells/pathology , Immediate-Early Proteins/genetics , Neoplasms/prevention & control , Polyploidy , Protein Tyrosine Phosphatases/genetics , Secondary Prevention/methods , Animals , Antineoplastic Agents/pharmacology , Immediate-Early Proteins/pharmacology , Mice , Neoplasms/pathology , Protein Tyrosine Phosphatases/pharmacology
5.
Nat Commun ; 10(1): 2484, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31171773

ABSTRACT

Tumor-specific antibody drugs can serve as cancer therapy with minimal side effects. A humanized antibody, PRL3-zumab, specifically binds to an intracellular oncogenic phosphatase PRL3, which is frequently expressed in several cancers. Here we show that PRL3-zumab specifically inhibits PRL3+ cancer cells in vivo, but not in vitro. PRL3 antigens are detected on the cell surface and outer exosomal membranes, implying an 'inside-out' externalization of PRL3. PRL3-zumab binds to surface PRL3 in a manner consistent with that in classical antibody-dependent cell-mediated cytotoxicity or antibody-dependent cellular phagocytosis tumor elimination pathways, as PRL3-zumab requires an intact Fc region and host FcγII/III receptor engagement to recruit B cells, NK cells and macrophages to PRL3+ tumor microenvironments. PRL3 is overexpressed in 80.6% of 151 fresh-frozen tumor samples across 11 common cancers examined, but not in patient-matched normal tissues, thereby implicating PRL3 as a tumor-associated antigen. Targeting externalized PRL3 antigens with PRL3-zumab may represent a feasible approach for anti-tumor immunotherapy.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Hepatocellular/metabolism , Cytophagocytosis/drug effects , Hepatocytes/drug effects , Liver Neoplasms/metabolism , Neoplasm Proteins/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , Tumor Microenvironment/drug effects , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antigens, Neoplasm/metabolism , B-Lymphocytes , Cell Line, Tumor , Hep G2 Cells , Hepatocytes/metabolism , Humans , Immunotherapy , Killer Cells, Natural , Macrophages , Mice , Molecular Targeted Therapy , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplasms/metabolism , Oncogene Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptors, IgG , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
6.
JCI Insight ; 1(9): e87607, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27699276

ABSTRACT

Novel, tumor-specific drugs are urgently needed for a breakthrough in cancer therapy. Herein, we generated a first-in-class humanized antibody (PRL3-zumab) against PRL-3, an intracellular tumor-associated phosphatase upregulated in multiple human cancers, for unconventional cancer immunotherapies. We focused on gastric cancer (GC), wherein elevated PRL-3 mRNA levels significantly correlated with shortened overall survival of GC patients. PRL-3 protein was overexpressed in 85% of fresh-frozen clinical gastric tumor samples examined but not in patient-matched normal gastric tissues. Using human GC cell lines, we demonstrated that PRL3-zumab specifically blocked PRL-3+, but not PRL-3-, orthotopic gastric tumors. In this setting, PRL3-zumab had better therapeutic efficacy as a monotherapy, rather than simultaneous combination with 5-fluorouracil or 5-fluorouracil alone. PRL3-zumab could also prevent PRL-3+ tumor recurrence. Mechanistically, we found that intracellular PRL-3 antigens could be externalized to become "extracellular oncotargets" that serve as bait for PRL3-zumab binding to potentially bridge and recruit immunocytes into tumor microenvironments for killing effects on cancer cells. In summary, our results document a comprehensive cancer therapeutic approach to specific antibody-targeted therapy against the PRL-3 oncotarget as a case study for developing antibodies against other intracellular targets in drug discovery.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Neoplasm Proteins/immunology , Protein Tyrosine Phosphatases/immunology , Stomach Neoplasms/therapy , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local , Xenograft Model Antitumor Assays
7.
Sci Rep ; 5: 17046, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26597054

ABSTRACT

PRL-3, a metastasis-associated phosphatase, is known to exert its oncogenic functions through activation of PI3K/Akt, which is a key regulator of the rapamycin-sensitive mTOR complex 1 (mTORC1), but a coherent link between PRL-3 and activation of mTOR has not yet been formally demonstrated. We report a positive correlation between PRL-3 expression and mTOR phospho-activation in clinical tumour samples and mouse models of cancer and demonstrate that PRL-3 increased downstream signalling to the mTOR substrates, p70S6K and 4E-BP1, by increasing PI3K/Akt-mediated activation of Rheb-GTP via TSC2 suppression. We also show that PRL-3 increases mTOR translocation to lysosomes via increased mTOR binding affinity to Rag GTPases in an Akt-independent manner, demonstrating a previously undescribed mechanism of action for PRL-3. PRL-3 also enhanced matrix metalloproteinase-2 secretion and cellular invasiveness via activation of mTOR, attributes which were sensitive to rapamycin treatment. The downstream effects of PRL-3 were maintained even under conditions of environmental stress, suggesting that PRL-3 provides a strategic survival advantage to tumour cells via its effects on mTOR.


Subject(s)
Multiprotein Complexes/metabolism , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Stomach Neoplasms/enzymology , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Movement , Disease Progression , Enzyme Activation , Humans , Lysosomes/enzymology , Matrix Metalloproteinase 2/metabolism , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Ras Homolog Enriched in Brain Protein , Signal Transduction , Stomach Neoplasms/pathology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
8.
Autophagy ; 10(10): 1787-800, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25136802

ABSTRACT

Autophagy, a "self-eating" cellular process, has dual roles in promoting and suppressing tumor growth, depending on cellular context. PTP4A3/PRL-3, a plasma membrane and endosomal phosphatase, promotes multiple oncogenic processes including cell proliferation, invasion, and cancer metastasis. In this study, we demonstrate that PTP4A3 accumulates in autophagosomes upon inhibition of autophagic degradation. Expression of PTP4A3 enhances PIK3C3-BECN1-dependent autophagosome formation and accelerates LC3-I to LC3-II conversion in an ATG5-dependent manner. PTP4A3 overexpression also enhances the degradation of SQSTM1, a key autophagy substrate. These functions of PTP4A3 are dependent on its catalytic activity and prenylation-dependent membrane association. These results suggest that PTP4A3 functions to promote canonical autophagy flux. Unexpectedly, following autophagy activation, PTP4A3 serves as a novel autophagic substrate, thereby establishing a negative feedback-loop that may be required to fine-tune autophagy activity. Functionally, PTP4A3 utilizes the autophagy pathway to promote cell growth, concomitant with the activation of AKT. Clinically, from the largest ovarian cancer data set (GSE 9899, n = 285) available in GEO, high levels of expression of both PTP4A3 and autophagy genes significantly predict poor prognosis of ovarian cancer patients. These studies reveal a critical role of autophagy in PTP4A3-driven cancer progression, suggesting that autophagy could be a potential Achilles heel to block PTP4A3-mediated tumor progression in stratified patients with high expression of both PTP4A3 and autophagy genes.


Subject(s)
Autophagy , Neoplasm Proteins/metabolism , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Protein Tyrosine Phosphatases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Biocatalysis/drug effects , CHO Cells , Cell Cycle Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Chloroquine/pharmacology , Cricetinae , Cricetulus , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Models, Biological , Ovarian Neoplasms/genetics , Phagosomes/drug effects , Phagosomes/metabolism , Prenylation/drug effects , Sequestosome-1 Protein , Substrate Specificity/drug effects , Survival Analysis
9.
EMBO Mol Med ; 5(9): 1351-66, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23929599

ABSTRACT

FLT3-ITD mutations are prevalent mutations in acute myeloid leukaemia (AML). PRL-3, a metastasis-associated phosphatase, is a downstream target of FLT3-ITD. This study investigates the regulation and function of PRL-3 in leukaemia cell lines and AML patients associated with FLT3-ITD mutations. PRL-3 expression is upregulated by the FLT3-STAT5 signalling pathway in leukaemia cells, leading an activation of AP-1 transcription factors via ERK and JNK pathways. PRL-3-depleted AML cells showed a significant decrease in cell growth. Clinically, high PRL-3 mRNA expression was associated with FLT3-ITD mutations in four independent AML datasets with 1158 patients. Multivariable Cox-regression analysis on our Cohort 1 with 221 patients identified PRL-3 as a novel prognostic marker independent of other clinical parameters. Kaplan-Meier analysis showed high PRL-3 mRNA expression was significantly associated with poorer survival among 491 patients with normal karyotype. Targeting PRL-3 reversed the oncogenic effects in FLT3-ITD AML models in vitro and in vivo. Herein, we suggest that PRL-3 could serve as a prognostic marker to predict poorer survival and as a promising novel therapeutic target for AML patients.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , fms-Like Tyrosine Kinase 3/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression , Humans
10.
J Clin Invest ; 123(8): 3459-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23867504

ABSTRACT

Metastasis-associated phosphatase of regenerating liver-3 (PRL-3) has pleiotropic effects in driving cancer progression, yet the signaling mechanisms of PRL-3 are still not fully understood. Here, we provide evidence for PRL-3-induced hyperactivation of EGFR and its downstream signaling cascades in multiple human cancer cell lines. Mechanistically, PRL-3-induced activation of EGFR was attributed primarily to transcriptional downregulation of protein tyrosine phosphatase 1B (PTP1B), an inhibitory phosphatase for EGFR. Functionally, PRL-3-induced hyperactivation of EGFR correlated with increased cell growth, promigratory characteristics, and tumorigenicity. Moreover, PRL-3 induced cellular addiction to EGFR signaling, as evidenced by the pronounced reversion of these oncogenic attributes upon EGFR-specific inhibition. Of clinical significance, we verified elevated PRL-3 expression as a predictive marker for favorable therapeutic response in a heterogeneous colorectal cancer (CRC) patient cohort treated with the clinically approved anti-EGFR antibody cetuximab. The identification of PRL-3-driven EGFR hyperactivation and consequential addiction to EGFR signaling opens new avenues for inhibiting PRL-3-driven cancer progression. We propose that elevated PRL-3 expression is an important clinical predictive biomarker for favorable anti-EGFR cancer therapy.


Subject(s)
Colorectal Neoplasms/metabolism , ErbB Receptors/metabolism , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Movement , Cell Proliferation , Cetuximab , Colorectal Neoplasms/drug therapy , Disease-Free Survival , Enzyme Repression , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Molecular Targeted Therapy , Phosphorylation , Protein Processing, Post-Translational , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction , Treatment Outcome , src-Family Kinases/metabolism
11.
BMC Cancer ; 13: 80, 2013 Feb 18.
Article in English | MEDLINE | ID: mdl-23418787

ABSTRACT

BACKGROUND: Phosphatase of regenerating liver-3 (PRL-3), a protein tyrosine phosphatase, is highly expressed in multiple human cancers and strongly implicated in tumor progression and cancer metastasis. However, the mechanisms by which PRL-3 promotes cancer cell migration, invasion, and metastasis are not very well understood. In this study, we investigated the contribution and molecular mechanisms of PRL-3 in ovarian cancer progression. METHODS: PRL-3 protein expression was detected on ovarian cancer tissue microarrays using immunohistochemistry. Stable PRL-3 depleted cell lines were generated using short hairpin RNA (shRNA) constructs. The migration and invasion potential of these cells were analyzed using Transwell and Matrigel assays, respectively. Immunoblotting and immunofluorescence were used to detect protein levels and distribution in PRL-3-ablated cells and the control cells. Cell morphology was observed with hematoxylin-eosin staining and transmission electron microscopy. Finally, PRL-3-ablated and control cells were injected into nude mice for xenograft tumorigenicity assays. RESULTS: Elevated PRL-3 expression was detected in 19% (26 out of 135) of human ovarian cancer patient samples, but not in normal ovary tissues (0 out of 14). Stable depletion of PRL-3 in A2780 ovarian cancer cells resulted in decreased migration ability and invasion activity compared with control parental A2780 cells. In addition, PRL-3-ablated cells also exhibited flattened morphology and extended lamellipodia. To address the possible molecular basis for the altered phenotypes associated with PRL-3 down-regulation, we assessed the expression profiles of various proteins involved in cell-matrix adhesion. Depletion of PRL-3 dramatically enhanced both RNA and protein levels of the cell surface receptor integrin α2, but not its heterologous binding partner integrin ß1. Inhibition of PRL-3 also correlated with elevated expression and phosphorylation of paxillin. A pronounced increase in the expression and activation of c-fos, a transcriptional activator of integrin α2, was observed in these PRL-3 knock-down cells. Moreover, forced expression of EGFP-PRL-3 resulted in the suppression of both integrin α2 and c-fos expression in A2780 cells. Significantly, using a xenograft tumor model, we observed a greatly reduced tumorigenicity of A2780 PRL-3 knock-down cells in vivo. CONCLUSIONS: These results suggest that PRL-3 plays a critical role in ovarian cancer tumorigenicity and maintaining the malignant phenotype. PRL-3 may inhibit c-fos transcriptional regulation of integrin α2 signaling. Our results strongly support a role for PRL-3 as a promising therapeutic target and potential early biomarker in ovarian cancer progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Integrin alpha2/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-fos/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Mice , Neoplasm Invasiveness , Ovarian Neoplasms/pathology , Paxillin/genetics , Transplantation, Heterologous , Tumor Burden/genetics , Up-Regulation/genetics
12.
Oncotarget ; 3(2): 158-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22374986

ABSTRACT

Antibodies are considered as 'magic bullets' because of their high specificity. It is believed that antibodies are too large to routinely enter the cytosol, thus antibody therapeutic approach has been limited to extracellular or secreted proteins expressed by cancer cells. However, many oncogenic proteins are localized within the cell. To explore the possibility of antibody therapies against intracellular targets, we generated a chimeric antibody targeting the intracellular PRL-3 oncoprotein to assess its antitumor activities in mice. Remarkably, we observed that the PRL-3 chimeric antibody could efficiently and specifically reduce the formation of PRL-3 expressing metastatic tumors. We further found that natural killer (NK) cells were important in mediating the therapeutic effect, which was only observed in a nude mouse model (T-cell deficient), but not in a Severe Combined Immunodeficiency' (scid ) mouse model (B- and T-cell deficient), indicating the anticancer effect also depends on host B-cell activity. Our study involving 377 nude and scid mice suggest that antibodies targeting intracellular proteins can be developed to treat cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Immediate-Early Proteins/immunology , Killer Cells, Natural/immunology , Molecular Targeted Therapy/methods , Protein Tyrosine Phosphatases/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Humans , Lymphocyte Activation/immunology , Melanoma/drug therapy , Mice , Mice, Nude , Mice, SCID , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
13.
Sci Transl Med ; 3(99): 99ra85, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21900592

ABSTRACT

Antibody-based therapies have better specificity and thus improved efficacy over standard chemotherapy regimens, which result in extended survival and improved quality of life for cancer patients. Because antibodies are viewed as too large to access intracellular locations, antibody therapy has traditionally targeted extracellular or secreted proteins expressed by cancer cells. However, many oncogenic proteins are found within the cell (such as intracellular phosphatases/kinases and transcription factors) and have therefore not been pursued for antibody therapies. Here, we explored the possibility of antibody therapy or vaccination against intracellular proteins. As proofs of concept, we selected three representative intracellular proteins as immunogens for tumor vaccine studies: PRL-3 (phosphatase of regenerating liver 3), a cancer-associated phosphatase; EGFP (enhanced green fluorescent protein), a general reporter; and mT (polyomavirus middle T), the polyomavirus middle T oncoprotein. A variety of tumors that expressed these intracellular proteins were clearly inhibited by their respective exogenous antibodies or by antigen-induced host antibodies (vaccination). These anticancer activities were reproducibly observed in hundreds of C57BL/6 tumor-bearing mice and MMTV-PymT transgenic breast tumor mice. Our in vivo data suggest that immunotherapies can target not only extracellular but also intracellular oncoproteins.


Subject(s)
Antineoplastic Agents/immunology , Cancer Vaccines/immunology , Immunotherapy/methods , Neoplasms/immunology , Oncogene Proteins/immunology , Vaccination , Animals , Antigens, Polyomavirus Transforming/immunology , Antineoplastic Agents/therapeutic use , B-Lymphocytes/immunology , Cell Line, Tumor , Female , Green Fluorescent Proteins/immunology , Humans , Immediate-Early Proteins/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Protein Tyrosine Phosphatases/immunology , Survival Rate
14.
J Cell Biochem ; 111(5): 1087-98, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21053359

ABSTRACT

The deregulated expression of members of the phosphatase of regenerating liver (PRL) family has been implicated in the metastatic progression of multiple human cancers. Importantly, PRL-1 and PRL-3 both possess the capacity to drive key steps in metastatic progression. Yet, little is known about the regulation and oncogenic mechanisms of this emerging class of dual-specificity phosphatases. This prospect article details the involvement of PRLs in the metastatic cascade, the regulatory mechanisms controlling PRL expression, and recent efforts in the characterization of PRL-modulated pathways and substrates using biochemical and high-throughput approaches. Current advances and future prospects in anti-cancer therapy targeting this family are also discussed.


Subject(s)
Neoplasm Metastasis/pathology , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Dual-Specificity Phosphatases , Humans , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...