1
|
Urasawa T, Kawasaki N. Proteomic Approach Using DIA-MS Identifies Morphogenesis-Associated Proteins during Cardiac Differentiation of Human iPS Cells. ACS OMEGA 2025; 10:344-357. [PMID: 39829588 PMCID: PMC11740111 DOI: 10.1021/acsomega.4c06371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes have potential applications in regenerative medicine. The quality by design (QbD) approach enables the efficiency and quality assurance in the manufacturing of hiPSC-derived products. It requires a molecular understanding of hiPSC differentiation throughout the differentiation process; however, information on cardiac differentiation remains limited. Proteins associated with the early stages of cardiac differentiation would be useful in the cardiomyocyte quality assessment. Here, we performed quantitative proteomics of hiPSC intermediate cells in the early phase of cardiac differentiation to better understand their molecular characteristics. Proteomic profiles suggested that day 5-7 cells were in the morphogenetic stage of cardiac differentiation. Trophoblast glycoprotein (TPBG) was the most up-regulated protein in the morphogenetic stage; it was previously shown to be up-regulated during differentiation into neural stem cells. Proteomics of TPBG-knockdown cells revealed that TPBG is involved in cell proliferation and is related to the cardiomyocyte yield, suggesting that it could be used as a marker in QbD development. Our approach helps us understand the molecular basis of hiPSC differentiation and could be a powerful tool in QbD-based manufacturing.
Collapse
Affiliation(s)
- Takaya Urasawa
- Biopharmaceutical and Regenerative
Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Nana Kawasaki
- Biopharmaceutical and Regenerative
Sciences, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
2
|
He Y, Tian R, Xu D, Wu Y, Rina S, Chen T, Guan Y, Xie T, Ying T, Xie F, Han J. Preclinical evaluation and pilot clinical study of [ 68Ga]Ga-NOTA-H006 for non-invasive PET imaging of 5T4 oncofetal antigen. Eur J Nucl Med Mol Imaging 2025; 52:611-622. [PMID: 39377811 DOI: 10.1007/s00259-024-06941-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Trophoblast glycoprotein, the so-called 5T4, is an oncofetal antigen expressed in many different cancers. However, no 5T4-specific radioligand is employed in the clinic for non-invasive diagnosis. Thus, the aim of the current study was to develop a PET radiotracer for imaging 5T4 expression in preclinical and clinical stages. METHODS A VHH library was constructed by camel immunization. The specificity of the VHHs toward 5T4 antigen was screened through phage display biopanning and periplasmic extract enzyme-linked immunosorbent assay. 1,4,7-Triazacyclononane-1,4,7-triacetate acid (NOTA) derivative was conjugated to the selected VHH. After radiolabeling, microPET/CT and ex vivo biodistribution were conducted using BxPC-3 and MDA-MB-468 tumor-bearing mice. Cold VHH was co-injected with the tracer to challenge its binding in vivo. For the pilot clinical study, PET/CT images were acquired at 1 h after injection of tracer in patients with pathologically confirmed primary and metastatic tumors. RESULTS A library with a capacity of 1.2 × 1012 colony-forming units was constructed after successful camel immunization. Nb1-40 with a median effect concentration of 0.43 nM was selected. After humanization, the resulting H006 maintained a high affinity towards 5T4. [68Ga]Ga-NOTA-H006 with the molar activities of 6.48-54.2 GBq/µmol was prepared with high radiochemical purity (> 98%). Using [68Ga]Ga-NOTA-H006, microPET/CT revealed a clear visualization of 5T4 expression in BxPC-3 tumor-bearing mice. Ex vivo biodistribution showed that the highest tumor-to-blood ratio (∼ 3-fold) and tumor-to-muscle ratio (∼ 5-fold) were achieved at 60 min post-injection. Co-injection of the cold H006 at a dose of 1.5 mg/kg significantly reduced the tumor uptake (p < 0.0001). In the pilot clinical study, [68Ga]Ga-NOTA-H006 demonstrated its capacity to map 5T4-positive lesions in humans and yielded a mean effective dose of 3.4 × 10- 2 mSv/MBq. CONCLUSIONS [68Ga]Ga-NOTA-H006, which can visualize 5T4 expression in vivo, has been successfully developed. This opens up opportunities for non-invasively studying 5T4 expression through nuclear medicine. Further clinical investigations are warranted to explore its clinical value in disease progression and companion diagnosis.
Collapse
Affiliation(s)
- Yingfang He
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Sa Rina
- Huahe Pharmaceutical Co., Ltd, Shanghai, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Tianwu Xie
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China.
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China.
| |
Collapse
|
3
|
Sun L, Shi M, Wang J, Han X, Wei J, Huang Z, Yang X, Ding Y, Zhang P, He A, Liu M, Yan R, Yang X, Li R, Wang G. Overexpressed Trophoblast Glycoprotein Contributes to Preeclampsia Development by Inducing Abnormal Trophoblast Migration and Invasion Toward the Uterine Spiral Artery. Hypertension 2024; 81:1524-1536. [PMID: 38716674 DOI: 10.1161/hypertensionaha.124.22923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Preeclampsia is a significant pregnancy disorder with an unknown cause, mainly attributed to impaired spiral arterial remodeling. METHODS Using RNA sequencing, we identified key genes in placental tissues from healthy individuals and preeclampsia patients. Placenta and plasma samples from pregnant women were collected to detect the expression of TPBG (trophoblast glycoprotein). Pregnant rats were injected with TPBG-carrying adenovirus to detect preeclamptic features. HTR-8/SVneo cells transfected with a TPBG overexpression lentiviral vector were used in cell function experiments. The downstream molecular mechanisms of TPBG were explored using RNA sequencing and single-cell RNA sequencing data. TPBG expression was knocked down in the lipopolysaccharide-induced preeclampsia-like rat model to rescue the preeclampsia features. We also assessed TPBG's potential as an early preeclampsia predictor using clinical plasma samples. RESULTS TPBG emerged as a crucial differentially expressed gene, expressed specifically in syncytiotrophoblasts and extravillous trophoblasts. Subsequently, we established a rat model with preeclampsia-like phenotypes by intravenously injecting TPBG-expressing adenoviruses, observing impaired spiral arterial remodeling, thus indicating a causal correlation between TPBG overexpression and preeclampsia. Studies with HTR-8/SVneo cells, chorionic villous explants, and transwell assays showed TPBG overexpression disrupts trophoblast/extravillous trophoblast migration/invasion and chemotaxis. Notably, TPBG knockdown alleviated the lipopolysaccharide-induced preeclampsia-like rat model. We enhanced preeclampsia risk prediction in early gestation by combining TPBG expression with established clinical predictors. CONCLUSIONS These findings are the first to show that TPBG overexpression contributes to preeclampsia development by affecting uterine spiral artery remodeling. We propose TPBG levels in maternal blood as a predictor of preeclampsia risk. The proposed mechanism by which TPBG overexpression contributes to the occurrence of preeclampsia via its disruptive effect on trophoblast and extravillous trophoblast migration/invasion on uterine spiral artery remodeling, thereby increasing the risk of preeclampsia.
Collapse
Affiliation(s)
- Lu Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Meiting Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Jian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xiaoxue Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Jiachun Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Zhengrui Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xiaofeng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
| | - Yuzhen Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Ping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
| | - Andong He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Mengyuan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Ruiling Yan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education (X.Y., G.W.), Jinan University, Guangzhou, China
- Clinical Research Center, Clifford Hospital, Guangzhou, China (X.Y.)
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University (L.S., M.S., J.W., X.H., J.W., Z.H., X.Y., Y.D., P.Z., A.H., M.L., R.Y., R.L.), Jinan University, Guangzhou, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine (P.Z., X.Y., G.W.), Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education (X.Y., G.W.), Jinan University, Guangzhou, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, School of Medicine (G.W.), Jinan University, Guangzhou, China
| |
Collapse
|
4
|
Kemper K, Gielen E, Boross P, Houtkamp M, Plantinga TS, de Poot SAH, Burm SM, Janmaat ML, Koopman LA, van den Brink EN, Rademaker R, Verzijl D, Engelberts PJ, Satijn D, Sasser AK, Breij ECW. Mechanistic and pharmacodynamic studies of DuoBody-CD3x5T4 in preclinical tumor models. Life Sci Alliance 2022; 5:5/11/e202201481. [PMID: 36271507 PMCID: PMC9458754 DOI: 10.26508/lsa.202201481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4− but not of 5T4−IFNGR1− tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.
Collapse
|
5
|
Ceci C, Lacal PM, Graziani G. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential. Pharmacol Ther 2022; 236:108106. [PMID: 34990642 DOI: 10.1016/j.pharmthera.2021.108106] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
6
|
Trophoblast glycoprotein is a new candidate gene for Parkinson’s disease. NPJ Parkinsons Dis 2021; 7:110. [PMID: 34876581 PMCID: PMC8651753 DOI: 10.1038/s41531-021-00252-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 11/04/2021] [Indexed: 11/08/2022] Open
Abstract
AbstractParkinson’s disease (PD) is a movement disorder caused by progressive degeneration of the midbrain dopaminergic (mDA) neurons in the substantia nigra pars compacta (SNc). Despite intense research efforts over the past decades, the etiology of PD remains largely unknown. Here, we discovered the involvement of trophoblast glycoprotein (Tpbg) in the development of PD-like phenotypes in mice. Tpbg expression was detected in the ventral midbrain during embryonic development and in mDA neurons in adulthood. Genetic ablation of Tpbg resulted in mild degeneration of mDA neurons in aged mice (12–14 months) with behavioral deficits reminiscent of PD symptoms. Through in silico analysis, we predicted potential TPBG-interacting partners whose functions were relevant to PD pathogenesis; this result was substantiated by transcriptomic analysis of the SNc of aged Tpbg knockout mice. These findings suggest that Tpbg is a new candidate gene associated with PD and provide a new insight into PD pathogenesis.
Collapse
|
7
|
Bansal D, Reimers MA, Knoche EM, Pachynski RK. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13020334. [PMID: 33477569 PMCID: PMC7831137 DOI: 10.3390/cancers13020334] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Although most prostate cancers are localized, and the majority are curable, recurrences occur in approximately 35% of men. Among patients with prostate-specific antigen (PSA) recurrence and PSA doubling time (PSADT) less than 15 months after radical prostatectomy, prostate cancer accounted for approximately 90% of the deaths by 15 years after recurrence. An immunosuppressive tumor microenvironment (TME) and impaired cellular immunity are likely largely responsible for the limited utility of checkpoint inhibitors (CPIs) in advanced prostate cancer compared with other tumor types. Thus, for immunologically "cold" malignancies such as prostate cancer, clinical trial development has pivoted towards novel approaches to enhance immune responses. Numerous clinical trials are currently evaluating combination immunomodulatory strategies incorporating vaccine-based therapies, checkpoint inhibitors, and chimeric antigen receptor (CAR) T cells. Other trials evaluate the efficacy and safety of these immunomodulatory agents' combinations with standard approaches such as androgen deprivation therapy (ADT), taxane-based chemotherapy, radiotherapy, and targeted therapies such as tyrosine kinase inhibitors (TKI) and poly ADP ribose polymerase (PARP) inhibitors. Here, we will review promising immunotherapies in development and ongoing trials for metastatic castration-resistant prostate cancer (mCRPC). These novel trials will build on past experiences and promise to usher a new era to treat patients with mCRPC.
Collapse
|
8
|
Wakeham CM, Ren G, Morgans CW. Expression and distribution of trophoblast glycoprotein in the mouse retina. J Comp Neurol 2020; 528:1660-1671. [PMID: 31891182 DOI: 10.1002/cne.24850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 02/03/2023]
Abstract
We recently identified the leucine-rich repeat (LRR) adhesion protein, trophoblast glycoprotein (TPBG), as a novel PKCα-dependent phosphoprotein in retinal rod bipolar cells (RBCs). Since TPBG has not been thoroughly examined in the retina, this study characterizes the localization and expression patterns of TPBG in the developing and adult mouse retina using two antibodies, one against the N-terminal LRR domain and the other against the C-terminal PDZ-interacting motif. Both antibodies labeled RBC dendrites in the outer plexiform layer and axon terminals in the IPL, as well as a putative amacrine cell with their cell bodies in the inner nuclear layer (INL) and a dense layer in the middle of the inner plexiform layer (IPL). In live transfected HEK293 cells, TPBG was localized to the plasma membrane with the N-terminal LRR domain facing the extracellular space. TPBG immunofluorescence in RBCs was strongly altered by the loss of TRPM1 in the adult retina, with significantly less dendritic and axon terminal labeling in TRPM1 knockout compared to wild type, despite no change in total TPBG detected by immunoblotting. During retinal development, TPBG expression increases dramatically just prior to eye opening with a time course closely correlated with that of TRPM1 expression. In the retina, LRR proteins have been implicated in the development and maintenance of functional bipolar cell synapses, and TPBG may play a similar role in RBCs.
Collapse
Affiliation(s)
- Colin M Wakeham
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Gaoying Ren
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
9
|
Takahashi Y, Takahashi H, Stern PL, Kirita T, Tsuboi A. Expression of Oncofetal Antigen 5T4 in Murine Taste Papillae. Front Cell Neurosci 2019; 13:343. [PMID: 31417363 PMCID: PMC6685444 DOI: 10.3389/fncel.2019.00343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Multicellular taste buds located within taste papillae on the tongue mediate taste sensation. In taste papillae, taste bud cells (TBCs), such as taste receptor cells and taste precursor cells, and the surrounding lingual epithelium including epithelial progenitors (also called taste stem/progenitor cells) are maintained by continuous cell turnover throughout life. However, it remains unknown how the cells constituting taste buds proliferate and differentiate to maintain taste bud tissue. Based on in situ hybridization (ISH) screening, we demonstrated that the oncofetal antigen 5T4 (also known as trophoblast glycoprotein: TPBG) gene is expressed in the adult mouse tongue. Results: In immunohistochemistry of coronal tongue sections, 5T4 protein was detected at a low level exclusively in the basal part of the lingual epithelium in developing and adult mice, and at a high level particularly in foliate papillae and circumvallate papillae (CVPs). Furthermore, immunohistochemistry of the basal part of CVPs indicated that the proliferation marker PCNA (proliferating cell nuclear antigen) co-localized with 5T4. 5T4 was strongly expressed in Krt5+ epithelial progenitors and Shh+ taste precursor cells, but weakly in mature taste receptor cells. The number of proliferating cells in the CVP was higher in 5T4-knockout mice than in wild-type (WT) mice, while neither cell differentiation nor the size of taste buds differed between these two groups of mice. Notably, X-ray irradiation enhanced cell proliferation more in 5T4-knockout mice than in WT mice. Conclusion: Our results suggest that 5T4, expressed in epithelial progenitors (taste stem/progenitor cells), and taste precursor cells, may influence the maintenance of taste papillae under both normal and injury conditions.
Collapse
Affiliation(s)
- Yuka Takahashi
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Hiroo Takahashi
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, Japan
| | - Peter L. Stern
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Akio Tsuboi
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
- Laboratory for Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
10
|
Wakeham CM, Wilmarth PA, Cunliffe JM, Klimek JE, Ren G, David LL, Morgans CW. Identification of PKCα-dependent phosphoproteins in mouse retina. J Proteomics 2019; 206:103423. [PMID: 31255707 DOI: 10.1016/j.jprot.2019.103423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022]
Abstract
Adjusting to a wide range of light intensities is an essential feature of retinal rod bipolar cell (RBC) function. While persuasive evidence suggests this modulation involves phosphorylation by protein kinase C-alpha (PKCα), the targets of PKCα phosphorylation in the retina have not been identified. PKCα activity and phosphorylation in RBCs was examined by immunofluorescence confocal microscopy using a conformation-specific PKCα antibody and antibodies to phosphorylated PKC motifs. PKCα activity was dependent on light and expression of TRPM1, and RBC dendrites were the primary sites of light-dependent phosphorylation. PKCα-dependent retinal phosphoproteins were identified using a phosphoproteomics approach to compare total protein and phosphopeptide abundance between phorbol ester-treated wild type and PKCα knockout (PKCα-KO) mouse retinas. Phosphopeptide mass spectrometry identified over 1100 phosphopeptides in mouse retina, with 12 displaying significantly greater phosphorylation in WT compared to PKCα-KO samples. The differentially phosphorylated proteins fall into the following functional groups: cytoskeleton/trafficking (4 proteins), ECM/adhesion (2 proteins), signaling (2 proteins), transcriptional regulation (3 proteins), and homeostasis/metabolism (1 protein). Two strongly differentially expressed phosphoproteins, BORG4 and TPBG, were localized to the synaptic layers of the retina, and may play a role in PKCα-dependent modulation of RBC physiology. Data are available via ProteomeXchange with identifier PXD012906. SIGNIFICANCE: Retinal rod bipolar cells (RBCs), the second-order neurons of the mammalian rod visual pathway, are able to modulate their sensitivity to remain functional across a wide range of light intensities, from starlight to daylight. Evidence suggests that this modulation requires the serine/threonine kinase, PKCα, though the specific mechanism by which PKCα modulates RBC physiology is unknown. This study examined PKCα phosophorylation patterns in mouse rod bipolar cells and then used a phosphoproteomics approach to identify PKCα-dependent phosphoproteins in the mouse retina. A small number of retinal proteins showed significant PKCα-dependent phosphorylation, including BORG4 and TPBG, suggesting a potential contribution to PKCα-dependent modulation of RBC physiology.
Collapse
Affiliation(s)
- Colin M Wakeham
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jennifer M Cunliffe
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - John E Klimek
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - Gaoying Ren
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Larry L David
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA; Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Catherine W Morgans
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
11
|
Role of TPBG (Trophoblast Glycoprotein) Antigen in Human Pericyte Migratory and Angiogenic Activity. Arterioscler Thromb Vasc Biol 2019; 39:1113-1124. [DOI: 10.1161/atvbaha.119.312665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective—
To determine the role of the oncofetal protein TPBG (trophoblast glycoprotein) in normal vascular function and reparative vascularization.
Approach and Results—
Immunohistochemistry of human veins was used to show TPBG expression in vascular smooth muscle cells and adventitial pericyte-like cells (APCs). ELISA, Western blot, immunocytochemistry, and proximity ligation assays evidenced a hypoxia-dependent upregulation of TPBG in APCs not found in vascular smooth muscle cells or endothelial cells. This involves the transcriptional modulator CITED2 (Atypical chemokine receptor 3 CBP/p300-interacting transactivator with glutamic acid (E)/aspartic acid (D)-rich tail) and downstream activation of CXCL12 (chemokine [C-X-C motif] ligand-12) signaling through the CXCR7 (C-X-C chemokine receptor type 7) receptor and ERK1/2 (extracellular signal-regulated kinases 1/2). TPBG silencing by siRNA transfection downregulated CXCL12, CXCR7, and pERK (phospho Thr202/Tyr204 ERK1/2) and reduced the APC migratory and proangiogenic capacities. TPBG forced expression induced opposite effects, which were associated with the formation of CXCR7/CXCR4 (C-X-C chemokine receptor type 4) heterodimers and could be contrasted by CXCL12 and CXCR7 neutralization. In vivo Matrigel plug assays using APCs with or without TPBG silencing evidenced TPBG is essential for angiogenesis. Finally, in immunosuppressed mice with limb ischemia, intramuscular injection of TPBG-overexpressing APCs surpassed naïve APCs in enhancing perfusion recovery and reducing the rate of toe necrosis.
Conclusions—
TPBG orchestrates the migratory and angiogenic activities of pericytes through the activation of the CXCL12/CXCR7/pERK axis. This novel mechanism could be a relevant target for therapeutic improvement of reparative angiogenesis.
Collapse
|
12
|
Harrop R, O’Neill E, Stern PL. Cancer stem cell mobilization and therapeutic targeting of the 5T4 oncofetal antigen. Ther Adv Vaccines Immunother 2019; 7:2515135518821623. [PMID: 30719508 PMCID: PMC6348545 DOI: 10.1177/2515135518821623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/27/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer stem cells (CSCs) can act as the cellular drivers of tumors harnessing stem cell properties that contribute to tumorigenesis either as founder elements or by the gain of stem cell traits by the malignant cells. Thus, CSCs can self-renew and generate the cellular heterogeneity of tumors including a hierarchical organization similar to the normal tissue. While the principle tumor growth contribution is often from the non-CSC components, it is the ability of small numbers of CSCs to avoid the effects of therapeutic strategies that can contribute to recurrence after treatment. However, identifying and characterizing CSCs for therapeutic targeting is made more challenging by their cellular potency being influenced by a particular tissue niche or by the capacity of more committed cells to regain stem cell functions. This review discusses the properties of CSCs including the limitations of the available cell surface markers, the assays that document tumor initiation and clonogenicity, the roles of epithelial mesenchymal transition and molecular pathways such as Notch, Wnt, Hippo and Hedgehog. The ability to target and eliminate CSCs is thought to be critical in the search for curative cancer treatments. The oncofetal tumor-associated antigen 5T4 (TBGP) has been linked with CSC properties in several different malignancies. 5T4 has functional attributes that are relevant to the spread of tumors including through EMT, CXCR4/CXCL12, Wnt, and Hippo pathways which may all contribute through the mobilization of CSCs. There are several different immunotherapies targeting 5T4 in development including antibody-drug conjugates, antibody-targeted bacterial super-antigens, a Modified Vaccinia Ankara-basedvaccine and 5T4-directed chimeric antigen receptor T-cells. These immune therapies would have the advantage of targeting both the bulk tumor as well as mobilized CSC populations.
Collapse
Affiliation(s)
- Richard Harrop
- Oxford BioMedica plc, Windrush Court, Transport Way, Oxford, OX4 6LT, UK
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Peter L. Stern
- Division of Molecular & Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Matsuoka K, Kohara Y, Naoe Y, Watanabe A, Ito M, Ikeda K, Takeshita S. WAIF1 Is a Cell-Surface CTHRC1 Binding Protein Coupling Bone Resorption and Formation. J Bone Miner Res 2018; 33:1500-1512. [PMID: 29624737 DOI: 10.1002/jbmr.3436] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/25/2018] [Accepted: 03/29/2018] [Indexed: 11/08/2022]
Abstract
The osteoclast-derived collagen triple helix repeat containing 1 (CTHRC1) protein stimulates osteoblast differentiation, but the underlying mechanism remains unclear. Here, we identified Wnt-activated inhibitory factor 1 (WAIF1)/5T4 as a cell-surface protein binding CTHRC1. The WAIF1-encoding Trophoblast glycoprotein (Tpbg) gene, which is abundantly expressed in the brain and bone but not in other tissues, showed the same expression pattern as Cthrc1. Tpbg downregulation in marrow stromal cells reduced CTHRC1 binding and CTHRC1-stimulated alkaline phosphatase activity through PKCδ activation of MEK/ERK, suggesting a novel WAIF1/PKCδ/ERK pathway triggered by CTHRC1. Unexpectedly, osteoblast lineage-specific deletion of Tpbg downregulated Rankl expression in mouse bones and reduced both bone formation and resorption; importantly, it impaired bone mass recovery following RANKL-induced resorption, reproducing the phenotype of osteoclast-specific Cthrc1 deficiency. Thus, the binding of osteoclast-derived CTHRC1 to WAIF1 in stromal cells activates PKCδ-ERK osteoblastogenic signaling and serves as a key molecular link between bone resorption and formation during bone remodeling. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Kazuhiko Matsuoka
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yukihiro Kohara
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yoshinori Naoe
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Atsushi Watanabe
- Laboratory of Research Advancement, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Masako Ito
- Medical Work-Life-Balance Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Kyoji Ikeda
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Sunao Takeshita
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| |
Collapse
|
14
|
Wang R, Lai Q, Lu Y, Zhou Y, Tang L, Tao Y, Yao Y, Yu L, Liu Y, Wang Y, Zhang R, Jiang X, Gou L, Yang J. Expression of 5T4 extracellular domain fusion protein and preparation of anti-5T4 monoclonal antibody with high affinity and internalization efficiency. Protein Expr Purif 2018; 158:51-58. [PMID: 29981846 DOI: 10.1016/j.pep.2018.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/26/2018] [Accepted: 07/04/2018] [Indexed: 02/05/2023]
Abstract
5T4, a membrane protein, is overexpressed in many tumor tissues but rarely expressed in normal tissues. Here, CHO-5T4+ cells were generated and served as the antigen to immunize mice. Hybridoma techniques were employed to produce monoclonal antibodies (mAbs). The recombinant protein of human IgG Fc-fused extracellular domain of 5T4 (5T4 ECD-Fc) was obtained from transient expression in HEK293F cells. The fusion protein 5T4 ECD-Fc and CHO-5T4+ cells were respectively utilized to screen anti-5T4 antibodies that could bind to the native antigen. In preliminary screening, three hundred and fifty mAbs were obtained. Via surface plasmon resonance and flow cytometry screening, seven anti-5T4 mAbs stood out. Among them, H6 showed a high affinity (KD = 1.6 × 10-11 M) and internalization percentage (36% for 1 h and 80% for 4 h). The molecular weight and isoelectric point of H6 were determined by LC-MS and iCIEF. Moreover, the specific reactivity of H6 was demonstrated by western blotting, flow cytometry, and immunohistochemistry, respectively. In conclusion, we produced human recombinant protein of 5T4 extracellular domain and developed high-affinity internalizing monoclonal antibodies which may be applied in the 5T4-targeting ADC therapy and basic research.
Collapse
Affiliation(s)
- Ruixue Wang
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qinhuai Lai
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Lu
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhou
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
| | - Liangze Tang
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yiran Tao
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Research Center for Public Health and Preventive Medicine, West China School of Public Health and Healthy Food Evaluation Research Center, NO. 4 West China Teaching Hospital, Sichuan University, Chengdu, China; Guangdong Zhongsheng Pharmaceutical Co., Ltd., China
| | - Lin Yu
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Liu
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxi Wang
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ruirui Zhang
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohua Jiang
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lantu Gou
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinliang Yang
- Department of Biotherapy, Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Guangdong Zhongsheng Pharmaceutical Co., Ltd., China.
| |
Collapse
|
15
|
Harris JL, Dave K, Gorman J, Khanna KK. The breast cancer antigen 5T4 interacts with Rab11, and is a target and regulator of Rab11 mediated trafficking. Int J Biochem Cell Biol 2018; 99:28-37. [DOI: 10.1016/j.biocel.2018.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 01/19/2023]
|
16
|
Wang R, Lai Q, Tang L, Tao Y, Yao Y, Liu Y, Lu Y, Shen C, Lu R, Fan C, Zhang R, Wang Y, Yu L, Yang T, Wu Y, Peng Y, Wei X, Fu Y, Lai W, Gou L, Yang J. A novel 5T4-targeting antibody-drug conjugate H6-DM4 exhibits potent therapeutic efficacy in gastrointestinal tumor xenograft models. Am J Cancer Res 2018; 8:610-623. [PMID: 29736307 PMCID: PMC5934552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/25/2018] [Indexed: 06/08/2023] Open
Abstract
5T4, also named as trophoblast glycoprotein, is often upregulated in some cancer cells. Here, we demonstrated that 5T4 was highly expressed in gastric, colorectal, and pancreatic cancer, associated with significantly poor prognosis of gastrointestinal (GI) cancer patients. To search for new targeting drugs for GI cancer, we developed a novel anti-5T4 monoclonal antibody with high affinity and robust internalization ability and conjugated it to the potent microtubule inhibitor DM4 to produce conjugate H6-DM4. This antibody-drug conjugate (ADC) displayed significant cytotoxicity in a panel of GI cancer cell lines with IC50 values in the nanomolar range. H6-DM4 eradicated established GI tumor xenograft models at 2.5 mg/kg or 10 mg/kg without observable toxicity. Further, 5T4 was highly expressed in cancer-initiating cells (CICs) compared with non-CICs in colorectal cancer. In vitro and in vivo, treatment with H6-DM4 exhibited a powerful efficacy on colorectal CICs. Additionally, colorectal cancer cells resistant to platinum were effectively eliminated by H6-DM4. Taken together, our results showed 5T4-positive GI cancer cells, colorectal cancer-initiating cells, and platinum-resistant colorectal cancer cells were potently eliminated by H6-DM4, indicating H6-DM4 may be a potential candidate drug for GI cancer treatment.
Collapse
Affiliation(s)
- Ruixue Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
- Guangdong Zhongsheng Pharmaceutical Co., Ltd.China
| | - Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Liangze Tang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Yiran Tao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Yuqin Yao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
- Research Center for Public Health and Preventive Medicine, West China School of Public Health and Healthy Food Evaluation Research Center, NO. 4 West China Teaching Hospital, Sichuan UniversityChengdu, China
- Guangdong Zhongsheng Pharmaceutical Co., Ltd.China
| | - Yu Liu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Ying Lu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Chaoyong Shen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan UniversityChengdu, China
| | - Ran Lu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Chuanwen Fan
- Institute of Digestive Surgery, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Ruirui Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Lin Yu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Tinghan Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan UniversityChengdu, China
| | - Yangping Wu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Yujia Peng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Xian Wei
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
- Guangdong Zhongsheng Pharmaceutical Co., Ltd.China
- School of Basic Medical Sciences, Chengdu Medical CollegeChengdu, China
| | - Yuyin Fu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Weirong Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Lantu Gou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, China
- Guangdong Zhongsheng Pharmaceutical Co., Ltd.China
| |
Collapse
|
17
|
The 5T4 oncofetal glycoprotein does not act as a general organizer of the CXCL12 system in cancer cells. Exp Cell Res 2018; 364:175-183. [PMID: 29408206 DOI: 10.1016/j.yexcr.2018.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 01/07/2023]
Abstract
The chemokine, CXCL12, promotes cancer growth and metastasis through interaction with either CXCR4 and/or CXCR7. This tumor-specific organization of the CXCL12 system obscures current therapeutic approaches, aiming at the selective inactivation of CXCL12 receptors. Since it has been previously suggested that the cellular use of CXCR4 or CXCR7 is dictated by the 5T4 oncofetal glycoprotein, we have now tested whether 5T4 would represent a general and reliable marker for the organization of the CXCL12 system in cancer cells. The CXCR4 antagonist, AMD3100, as well as the CXCR7 antagonist, CCX771, demonstrated that the cancer cell lines A549, C33A, DLD-1, MDA-231, and PC-3 use either CXCR7 and/or CXCR4 for mediating CXCL12-induced chemotaxis and cell proliferation. The use of CXCL12 receptors as well as their subcellular localization remained unchanged in most cell lines following siRNA-mediated depletion of 5T4. In distinct cell lines, inhibition of 5T4 expression, however, modulated tumor cell migration and proliferation per se. Collectively our analyses fail to demonstrate general organizational influences of 5T4 of the CXCL12 system in different cancer cell lines, and, hence, dismiss its future use as a diagnostic marker.
Collapse
|
18
|
Jiang Z, Li Y, Ji X, Tang Y, Yu H, Ding L, Yu M, Cui Q, Zhang M, Ma Y, Li M. Protein profiling identified key chemokines that regulate the maintenance of human pluripotent stem cells. Sci Rep 2017; 7:14510. [PMID: 29109449 PMCID: PMC5674019 DOI: 10.1038/s41598-017-15081-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 10/20/2017] [Indexed: 12/20/2022] Open
Abstract
Microenvironment (or niche)-providing chemokines regulate many important biological functions of tissue-specific stem cells. However, to what extent chemokines influence human pluripotent stem cells (hPSCs) is not yet completely understood. In this study, we applied protein array to screen chemokines found within the cytokine pool in the culture supernatant of hPSCs. Our results showed that chemokines were the predominant supernatant components, and came from three sources: hPSCs, feeder cells, and culture media. Chemotaxis analysis of IL-8, SDF-1α, and IP-10 suggested that chemokines function as uniform chemoattractants to mediate in vitro migration of the hPSCs. Chemokines mediate both differentiated and undifferentiated states of hPSCs. However, balanced chemokine signaling tends to enhance their stemness in vitro. These results indicate that chemokines secreted from both stem cells and feeder cells are essential to mobilize hPSCs and maintain their stemness.
Collapse
Affiliation(s)
- Zongmin Jiang
- Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China.,Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China
| | - Yonggang Li
- Department of Reproduction and Genetics, the First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
| | - Xinglai Ji
- Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan, 650091, China
| | - Yiyuli Tang
- Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan, 650091, China
| | - Haijing Yu
- Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China.,Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China
| | - Lei Ding
- Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China.,Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China
| | - Min Yu
- Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China.,Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China
| | - Qinghua Cui
- Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China.,Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China
| | - Ming Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yanping Ma
- Department of Reproduction and Genetics, the First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China.
| | - Meizhang Li
- Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China. .,Key Laboratory of Molecular Cancer Biology, Yunnan Education Department, Kunming, Yunnan, 650091, China.
| |
Collapse
|
19
|
A Subtype of Olfactory Bulb Interneurons Is Required for Odor Detection and Discrimination Behaviors. J Neurosci 2017; 36:8210-27. [PMID: 27488640 DOI: 10.1523/jneurosci.2783-15.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/04/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED Neural circuits that undergo reorganization by newborn interneurons in the olfactory bulb (OB) are necessary for odor detection and discrimination, olfactory memory, and innate olfactory responses, including predator avoidance and sexual behaviors. The OB possesses many interneurons, including various types of granule cells (GCs); however, the contribution that each type of interneuron makes to olfactory behavioral control remains unknown. Here, we investigated the in vivo functional role of oncofetal trophoblast glycoprotein 5T4, a regulator for dendritic arborization of 5T4-expressing GCs (5T4 GCs), the level of which is reduced in the OB of 5T4 knock-out (KO) mice. Electrophysiological recordings with acute OB slices indicated that external tufted cells (ETCs) can be divided into two types, bursting and nonbursting. Optogenetic stimulation of 5T4 GCs revealed their connection to both bursting and nonbursting ETCs, as well as to mitral cells (MCs). Interestingly, nonbursting ETCs received fewer inhibitory inputs from GCs in 5T4 KO mice than from those in wild-type (WT) mice, whereas bursting ETCs and MCs received similar inputs in both mice. Furthermore, 5T4 GCs received significantly fewer excitatory inputs in 5T4 KO mice. Remarkably, in olfactory behavior tests, 5T4 KO mice had higher odor detection thresholds than the WT, as well as defects in odor discrimination learning. Therefore, the loss of 5T4 attenuates inhibitory inputs from 5T4 GCs to nonbursting ETCs and excitatory inputs to 5T4 GCs, contributing to disturbances in olfactory behavior. Our novel findings suggest that, among the various types of OB interneurons, the 5T4 GC subtype is required for odor detection and discrimination behaviors. SIGNIFICANCE STATEMENT Neuronal circuits in the brain include glutamatergic principal neurons and GABAergic interneurons. Although the latter is a minority cell type, they are vital for normal brain function because they regulate the activity of principal neurons. If interneuron function is impaired, brain function may be damaged, leading to behavior disorder. The olfactory bulb (OB) possesses various types of interneurons, including granule cells (GCs); however, the contribution that each type of interneuron makes to the control of olfactory behavior remains unknown. Here, we analyzed electrophysiologically and behaviorally the function of oncofetal trophoblast glycoprotein 5T4, a regulator for dendritic branching in OB GCs. We found that, among the various types of OB interneuron, the 5T4 GC subtype is required for odor detection and odor discrimination behaviors.
Collapse
|
20
|
Harper J, Lloyd C, Dimasi N, Toader D, Marwood R, Lewis L, Bannister D, Jovanovic J, Fleming R, D'Hooge F, Mao S, Marrero AM, Korade M, Strout P, Xu L, Chen C, Wetzel L, Breen S, van Vlerken-Ysla L, Jalla S, Rebelatto M, Zhong H, Hurt EM, Hinrichs MJ, Huang K, Howard PW, Tice DA, Hollingsworth RE, Herbst R, Kamal A. Preclinical Evaluation of MEDI0641, a Pyrrolobenzodiazepine-Conjugated Antibody-Drug Conjugate Targeting 5T4. Mol Cancer Ther 2017; 16:1576-1587. [PMID: 28522587 DOI: 10.1158/1535-7163.mct-16-0825] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/28/2017] [Accepted: 04/28/2017] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) are used to selectively deliver cytotoxic agents to tumors and have the potential for increased clinical benefit to cancer patients. 5T4 is an oncofetal antigen overexpressed on the cell surface in many carcinomas on both bulk tumor cells as well as cancer stem cells (CSC), has very limited normal tissue expression, and can internalize when bound by an antibody. An anti-5T4 antibody was identified and optimized for efficient binding and internalization in a target-specific manner, and engineered cysteines were incorporated into the molecule for site-specific conjugation. ADCs targeting 5T4 were constructed by site-specifically conjugating the antibody with payloads that possess different mechanisms of action, either a DNA cross-linking pyrrolobenzodiazepine (PBD) dimer or a microtubule-destabilizing tubulysin, so that each ADC had a drug:antibody ratio of 2. The resulting ADCs demonstrated significant target-dependent activity in vitro and in vivo; however, the ADC conjugated with a PBD payload (5T4-PBD) elicited more durable antitumor responses in vivo than the tubulysin conjugate in xenograft models. Likewise, the 5T4-PBD more potently inhibited the growth of 5T4-positive CSCs in vivo, which likely contributed to its superior antitumor activity. Given that the 5T4-PBD possessed both potent antitumor activity as well as anti-CSC activity, and thus could potentially target bulk tumor cells and CSCs in target-positive indications, it was further evaluated in non-GLP rat toxicology studies that demonstrated excellent in vivo stability with an acceptable safety profile. Taken together, these preclinical data support further development of 5T4-PBD, also known as MEDI0641, against 5T4+ cancer indications. Mol Cancer Ther; 16(8); 1576-87. ©2017 AACR.
Collapse
Affiliation(s)
- Jay Harper
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland.
| | - Christopher Lloyd
- Antibody Discovery and Protein Engineering, MedImmune, Ltd, Cambridge, United Kingdom
| | - Nazzareno Dimasi
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland
| | - Dorin Toader
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland
| | - Rose Marwood
- Antibody Discovery and Protein Engineering, MedImmune, Ltd, Cambridge, United Kingdom
| | - Leeanne Lewis
- Antibody Discovery and Protein Engineering, MedImmune, Ltd, Cambridge, United Kingdom
| | - David Bannister
- Antibody Discovery and Protein Engineering, MedImmune, Ltd, Cambridge, United Kingdom
| | - Jelena Jovanovic
- Antibody Discovery and Protein Engineering, MedImmune, Ltd, Cambridge, United Kingdom
| | - Ryan Fleming
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland
| | | | - Shenlan Mao
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | | | - Martin Korade
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | - Patrick Strout
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | - Linda Xu
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland
| | - Cui Chen
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | - Leslie Wetzel
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | - Shannon Breen
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | | | - Sanjoo Jalla
- Project Management, MedImmune, LLC, Gaithersburg, Maryland
| | | | - Haihong Zhong
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | - Elaine M Hurt
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | | | - Keven Huang
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | | | - David A Tice
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | | | - Ronald Herbst
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland
| | - Adeela Kamal
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland.,Ferring Pharmaceuticals, San Diego, California
| |
Collapse
|
21
|
Alam SMK, Jasti S, Kshirsagar SK, Tannetta DS, Dragovic RA, Redman CW, Sargent IL, Hodes HC, Nauser TL, Fortes T, Filler AM, Behan K, Martin DR, Fields TA, Petroff BK, Petroff MG. Trophoblast Glycoprotein (TPGB/5T4) in Human Placenta: Expression, Regulation, and Presence in Extracellular Microvesicles and Exosomes. Reprod Sci 2017; 25:185-197. [PMID: 28481180 DOI: 10.1177/1933719117707053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Many parallels exist between growth and development of the placenta and that of cancer. One parallel is shared expression of antigens that may have functional importance and may be recognized by the immune system. Here, we characterize expression and regulation of one such antigen, Trophoblast glycoprotein (TPGB; also called 5T4), in the placenta across gestation, in placentas of preeclamptic (PE) pregnancies, and in purified microvesicles and exosomes. METHODS Trophoblast glycoprotein expression was analyzed by real-time reverse transcription-polymerase chain reaction (RT-PCR), Western blot, and immunohistochemistry. Regulation of 5T4 in cytotrophoblast cells was examined under either differentiating conditions of epidermal growth factor or under varying oxygen conditions. Microvesicles and exosomes were purified from supernatant of cultured and perfused placentas. RESULTS Trophoblast glycoprotein expression was prominent at the microvillus surface of syncytiotrophoblast and on the extravillous trophoblast cells, with minimal expression in undifferentiated cytotrophoblasts and normal tissues. Trophoblast glycoprotein expression was elevated in malignant tumors. In cytotrophoblasts, 5T4 was induced by in vitro differentiation, and its messenger RNA (mRNA) was increased under conditions of low oxygen. PE placentas expressed higher 5T4 mRNA than matched control placentas. Trophoblast glycoprotein was prominent within shed placental microvesicles and exosomes. CONCLUSION Given the potential functional and known immunological importance of 5T4 in cancer, these studies reveal a class of proteins that may influence placental development and/or sensitize the maternal immune system. In extravillous trophoblasts, 5T4 may function in epithelial-to-mesenchymal transition during placentation. The role of syncytiotrophoblast 5T4 is unknown, but its abundance in shed syncytial vesicles may signify route of sensitization of the maternal immune system.
Collapse
Affiliation(s)
- S M K Alam
- 1 Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.,2 Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - S Jasti
- 1 Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - S K Kshirsagar
- 3 Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - D S Tannetta
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - R A Dragovic
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - C W Redman
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - I L Sargent
- 4 Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - H C Hodes
- 5 Center for Women's Health, Overland Park, KS, USA
| | - T L Nauser
- 5 Center for Women's Health, Overland Park, KS, USA
| | - T Fortes
- 6 Sparrow Hospital, Lansing, MI, USA.,7 College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - A M Filler
- 6 Sparrow Hospital, Lansing, MI, USA.,7 College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - K Behan
- 7 College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | | | - T A Fields
- 8 Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - B K Petroff
- 3 Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA.,9 Veterinary Diagnostic Laboratory, Michigan State University, East Lansing, MI, USA
| | - M G Petroff
- 1 Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.,3 Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA.,10 Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
22
|
Stern PL, Harrop R. 5T4 oncofoetal antigen: an attractive target for immune intervention in cancer. Cancer Immunol Immunother 2017; 66:415-426. [PMID: 27757559 PMCID: PMC11029567 DOI: 10.1007/s00262-016-1917-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/12/2016] [Indexed: 01/18/2023]
Abstract
The natural history of a patient's cancer is often characterised by genetic diversity and sequential sweeps of clonal dominance. It is therefore not surprising that identifying the most appropriate tumour-associated antigen for targeted intervention is challenging. The 5T4 oncofoetal antigen was identified by searching for surface molecules shared between human trophoblast and cancer cells with the rationale that they may function to allow survival of the foetus as a semi-allograft in the mother or a tumour in its host. The 5T4 protein is expressed by many different cancers but rarely in normal adult tissues. 5T4 molecules are 72 kD, heavily N-glycosylated proteins with several leucine-rich repeats which are often associated with protein-protein interactions. 5T4 expression is associated with the directional movement of cells through epithelial mesenchymal transition, potentiation of CXCL12/CXCR4 chemotaxis and inhibition of canonical Wnt/beta-catenin while favouring non-canonical pathway signalling; all processes which help drive the spread of cancer cells. The selective pattern of 5T4 tumour expression, association with a tumour-initiating phenotype plus a mechanistic involvement with cancer spread have underwritten the clinical development of different immunotherapeutic strategies including a vaccine, a tumour-targeted superantigen and an antibody drug conjugate. In addition, a chimeric antigen receptor T cell approach targeting 5T4 expressing tumour cells is in pre-clinical development. A key challenge will include how best to combine each 5T4 targeted immunotherapy with the most appropriate standard of care treatment (or adjunct therapy) to maximise the recovery of immune control and ultimately eliminate the tumour.
Collapse
Affiliation(s)
- Peter L Stern
- Institute of Cancer Studies, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | - Richard Harrop
- Oxford BioMedica Plc, Windrush Court, Transport Way, Oxford, OX4 6LT, UK.
| |
Collapse
|
23
|
McGinn OJ, Krishnan S, Bourquin JP, Sapra P, Dempsey C, Saha V, Stern PL. Targeting the 5T4 oncofetal glycoprotein with an antibody drug conjugate (A1mcMMAF) improves survival in patient-derived xenograft models of acute lymphoblastic leukemia. Haematologica 2017; 102:1075-1084. [PMID: 28341731 PMCID: PMC5451339 DOI: 10.3324/haematol.2016.158485] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 03/15/2017] [Indexed: 12/29/2022] Open
Abstract
Outcome in childhood acute lymphoblastic leukemia is prognosticated from levels of minimal residual disease after remission induction therapy. Higher levels of minimal residual disease are associated with inferior results even with intensification of therapy, thus suggesting that identification and targeting of minimal residual disease cells could be a therapeutic strategy. Here we identify high expression of 5T4 in subclonal populations of patient-derived xenografts from patients with high, post-induction levels of minimal residual disease. 5T4-positive cells showed preferential ability to overcome the NOD-scidIL2Rγnull mouse xenograft barrier, migrated in vitro on a CXCL12 gradient, preferentially localized to bone marrow in vivo and displayed the ability to reconstitute the original clonal composition on limited dilution engraftment. Treatment with A1mcMMAF (a 5T4-antibody drug conjugate) significantly improved survival without overt toxicity in mice engrafted with a 5T4-positive acute lymphoblastic leukemia cell line. Mice engrafted with 5T4-positive patient-derived xenograft cells were treated with combination chemotherapy or dexamethasone alone and then given A1mcMMAF in the minimal residual disease setting. Combination chemotherapy was toxic to NOD-scidIL2Rγnull mice. While dexamethasone or A1mcMMAF alone improved outcomes, the sequential administration of dexamethasone and A1mcMMAF significantly improved survival (P=0.0006) over either monotherapy. These data show that specifically targeting minimal residual disease cells improved outcomes and support further investigation of A1mcMMAF in patients with high-risk B-cell precursor acute lymphoblastic leukemia identified by 5T4 expression at diagnosis.
Collapse
Affiliation(s)
- Owen J McGinn
- Immunology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK
| | - Shekhar Krishnan
- Paediatric Oncology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK.,Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, India
| | - Jean-Pierre Bourquin
- Division of Oncology & Children's Research Center, University Children's Hospital, University of Zurich, Switzerland
| | - Puja Sapra
- Pfizer Inc. Pearl River, NY10965-1299, USA
| | - Clare Dempsey
- Paediatric Oncology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK
| | - Vaskar Saha
- Paediatric Oncology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK .,Tata Translational Cancer Research Center, Tata Medical Center, Kolkata, India
| | - Peter L Stern
- Immunology, Division of Molecular & Clinical Cancer Sciences, University of Manchester, UK
| |
Collapse
|
24
|
Takahashi H, Tsuboi A. Olfactory Habituation-dishabituation Test (Mouse). Bio Protoc 2017; 7:e2154. [PMID: 34458470 DOI: 10.21769/bioprotoc.2154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 11/02/2022] Open
Abstract
Olfaction plays a fundamental role in the various behaviors such as feeding, mating, nursing, and avoidance in mice. Behavioral tests that characterize abilities of odor detection and recognition using genetically modified mice reveal the contribution of target genes to the olfactory processing. Here, we describe the olfactory habituation-dishabituation test for investigating the odor detection threshold in mice.
Collapse
Affiliation(s)
- Hiroo Takahashi
- Laboratory for Molecular Biology of Neural System, Advanced Medical Research Center, Nara Medical University, Kashihara, Nara, Japan
| | - Akio Tsuboi
- Laboratory for Molecular Biology of Neural System, Advanced Medical Research Center, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
25
|
Kerk SA, Finkel KA, Pearson AT, Warner KA, Zhang Z, Nör F, Wagner VP, Vargas PA, Wicha MS, Hurt EM, Hollingsworth RE, Tice DA, Nör JE. 5T4-Targeted Therapy Ablates Cancer Stem Cells and Prevents Recurrence of Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2016; 23:2516-2527. [PMID: 27780858 DOI: 10.1158/1078-0432.ccr-16-1834] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/06/2016] [Accepted: 10/10/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Locoregional recurrence is a frequent treatment outcome for patients with advanced head and neck squamous cell carcinoma (HNSCC). Emerging evidence suggests that tumor recurrence is mediated by a small subpopulation of uniquely tumorigenic cells, that is, cancer stem cells (CSC), that are resistant to conventional chemotherapy, endowed with self-renewal and multipotency.Experimental Design: Here, we evaluated the efficacy of MEDI0641, a novel antibody-drug conjugate targeted to 5T4 and carrying a DNA-damaging "payload" (pyrrolobenzodiazepine) in preclinical models of HNSCC.Results: Analysis of a tissue microarray containing 77 HNSCC with follow-up of up to 12 years revealed that patients with 5T4high tumors displayed lower overall survival than those with 5T4low tumors (P = 0.038). 5T4 is more highly expressed in head and neck CSC (ALDHhighCD44high) than in control cells (non-CSC). Treatment with MEDI0641 caused a significant reduction in the CSC fraction in HNSCC cells (UM-SCC-11B, UM-SCC-22B) in vitro Notably, a single intravenous dose of 1 mg/kg MEDI0641 caused long-lasting tumor regression in three patient-derived xenograft (PDX) models of HNSCC. MEDI0641 ablated CSC in the PDX-SCC-M0 model, reduced it by five-fold in the PDX-SCC-M1, and two-fold in the PDX-SCC-M11 model. Importantly, mice (n = 12) treated with neoadjuvant, single administration of MEDI0641 prior to surgical tumor removal showed no recurrence for more than 200 days, whereas the control group had 7 recurrences (in 12 mice; P = 0.0047).Conclusions: Collectively, these findings demonstrate that an anti-5T4 antibody-drug conjugate reduces the fraction of CSCs and prevents local recurrence and suggest a novel therapeutic approach for patients with HNSCC. Clin Cancer Res; 23(10); 2516-27. ©2016 AACR.
Collapse
Affiliation(s)
- Samuel A Kerk
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan
| | - Kelsey A Finkel
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan
| | - Alexander T Pearson
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan.,Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Kristy A Warner
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan
| | - Zhaocheng Zhang
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan
| | - Felipe Nör
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan.,Department of Oral Pathology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Vivian P Wagner
- Department of Oral Pathology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan
| | - Pablo A Vargas
- Department of Oral Diagnosis, University of Campinas, Piracicaba, Brazil
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Elaine M Hurt
- Oncology Research, MedImmune, Gaithersburg, Maryland
| | | | - David A Tice
- Oncology Research, MedImmune, Gaithersburg, Maryland
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan. .,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan.,Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
| |
Collapse
|
26
|
A gonogenic stimulated transition of mouse embryonic stem cells with enhanced control of diverse differentiation pathways. Sci Rep 2016; 6:25104. [PMID: 27157261 PMCID: PMC4860597 DOI: 10.1038/srep25104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/08/2016] [Indexed: 11/17/2022] Open
Abstract
Embryonic stem (ES) cells share markers with undifferentiated primordial germ cells (PGCs). Here, we discovered that a cellular state with some molecular markers of male gonocyte induction, including a G1/S phase arrest and upregulation of specific genes such as Nanos2, Tdrd1, Ddx4, Zbtb16 and Plk1s1, can be chemically induced in male mouse ES cells in vitro, which we termed gonogenic stimulated transition (GoST). After longer culture of the resulting GoST cells without chemical stimulation, several molecular markers typical for early gonocytes were detected including the early gonocyte marker Tex101. Motivated by previous studies that found multipotency in cell lines derived from neonatal male germ cells in vitro, we then compared the differentiation potential of GoST cells to that of ES cells in vitro. Interestingly, GoST cells showed equal neurogenic, but enhanced cardiogenic and hepatogenic differentiation compared to ES cells in vitro. This work shows for the first time that some important molecular markers of the first developmental sexual differentiation program can be induced in male mouse ES cells in vitro and defines a novel concept to generate cells with enhanced multipotency.
Collapse
|
27
|
Choi HY, Saha SK, Kim K, Kim S, Yang GM, Kim B, Kim JH, Cho SG. G protein-coupled receptors in stem cell maintenance and somatic reprogramming to pluripotent or cancer stem cells. BMB Rep 2015; 48:68-80. [PMID: 25413305 PMCID: PMC4352616 DOI: 10.5483/bmbrep.2015.48.2.250] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are a large class of transmembrane receptors categorized into five distinct families: rhodopsin, secretin, adhesion, glutamate, and frizzled. They bind and regulate 80% of all hormones and account for 20-50% of the pharmaceuticals currently on the market. Hundreds of GPCRs integrate and coordinate the functions of individual cells, mediating signaling between various organs. GPCRs are crucial players in tumor progression, adipogenesis, and inflammation. Several studies have also confirmed their central roles in embryonic development and stem cell maintenance. Recently, GPCRs have emerged as key players in the regulation of cell survival, proliferation, migration, and self-renewal in pluripotent (PSCs) and cancer stem cells (CSCs). Our study and other reports have revealed that the expression of many GPCRs is modulated during the generation of induced PSCs (iPSCs) or CSCs as well as during CSC sphere formation. These GPCRs may have crucial roles in the regulation of selfrenewal and other biological properties of iPSCs and CSCs. This review addresses the current understanding of the role of GPCRs in stem cell maintenance and somatic reprogramming to PSCs or CSCs.
Collapse
Affiliation(s)
- Hye Yeon Choi
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| | - Subbroto Kumar Saha
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| | - Kyeongseok Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| | - Sangsu Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| | - Gwang-Mo Yang
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| | - BongWoo Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| | - Jin-hoi Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| | - Ssang-Goo Cho
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul 143-701, Korea
| |
Collapse
|
28
|
Evolving Strategies for Target Selection for Antibody-Drug Conjugates. Pharm Res 2015; 32:3494-507. [PMID: 25585957 DOI: 10.1007/s11095-015-1624-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 01/06/2015] [Indexed: 01/06/2023]
Abstract
Antibody-drug conjugates (ADCs) represent a promising modality for the treatment of cancer. The therapeutic strategy is to deliver a potent drug preferentially to the tumor and not normal tissues by attaching the drug to an antibody that recognizes a tumor antigen. The selection of antigen targets is critical to enabling a therapeutic window for the ADC and has proven to be surprisingly complex. We surveyed the tumor and normal tissue expression profiles of the targets of ADCs currently in clinical development. Our analysis demonstrates a surprisingly broad range of expression profiles and the inability to formalize any optimal parameters for an ADC target. In this context, we discuss additional considerations for ADC target selection, including interdependencies among biophysical properties of the drug, biological functions of the target and strategies for clinical development. The TPBG (5T4) oncofetal antigen and the anti-TPBG ADC A1-mcMMAF are highlighted to demonstrate the relevance of the target's biological function. Emerging platform technologies and novel biological insights are expanding ADC target space and transforming strategies for target selection.
Collapse
|
29
|
Hu G, Leal M, Lin Q, Affolter T, Sapra P, Bates B, Damelin M. Phenotype of TPBG Gene Replacement in the Mouse and Impact on the Pharmacokinetics of an Antibody-Drug Conjugate. Mol Pharm 2014; 12:1730-7. [PMID: 25423493 DOI: 10.1021/mp5006323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The use of predictive preclinical models in drug discovery is critical for compound selection, optimization, preclinical to clinical translation, and strategic decision-making. Trophoblast glycoprotein (TPBG), also known as 5T4, is the therapeutic target of several anticancer agents currently in clinical development, largely due to its high expression in tumors and low expression in normal adult tissues. In this study, mice were engineered to express human TPBG under endogenous regulatory sequences by replacement of the murine Tpbg coding sequence. The gene replacement was considered functional since the hTPBG knockin (hTPBG-KI) mice did not exhibit clinical observations or histopathological phenotypes that are associated with Tpbg gene deletion, except in rare instances. The expression of hTPBG in certain epithelial cell types and in different microregions of the brain and spinal cord was consistent with previously reported phenotypes and expression patterns. In pharmacokinetic studies, the exposure of a clinical-stage anti-TPBG antibody-drug conjugate (ADC), A1mcMMAF, was lower in hTPBG-KI versus wild-type animals, which was evidence of target-related increased clearance in hTPBG-KI mice. Thus, the hTPBG-KI mice constitute an improved system for pharmacology studies with current and future TPBG-targeted therapies and can generate more precise pharmacokinetic and pharmacodynamic data. In general the strategy of employing gene replacement to improve pharmacokinetic assessments should be broadly applicable to the discovery and development of ADCs and other biotherapeutics.
Collapse
Affiliation(s)
| | - Mauricio Leal
- §Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Pearl River, New York 10965, United States
| | - Qingcong Lin
- ∥Global Biotherapeutic Technologies, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | | | - Puja Sapra
- #Oncology Research Unit, Pfizer Inc., Pearl River, New York 10965, United States
| | - Brian Bates
- ∥Global Biotherapeutic Technologies, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Marc Damelin
- #Oncology Research Unit, Pfizer Inc., Pearl River, New York 10965, United States
| |
Collapse
|
30
|
Eisen T, Hedlund G, Forsberg G, Hawkins R. Naptumomab estafenatox: targeted immunotherapy with a novel immunotoxin. Curr Oncol Rep 2014; 16:370. [PMID: 24445502 PMCID: PMC3918406 DOI: 10.1007/s11912-013-0370-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Improvement of cancer therapy by introducing new concepts is still urgent even though there have been major advancements lately. Immunotherapy is well on the way to becoming an established tool in the cancer treatment armory. It seems that a combination of (1) activation of immune effector cells and selective targeting of them to tumors and (2) the inhibition of immune suppression often induced by the tumor itself are necessary to achieve the therapeutic goal. The immunotoxin naptumomab estafenatox was developed in an effort to activate and target the patient’s own T cells to their tumor, by fusing a superantigen (SAg) variant that activates T lymphocytes to the Fab moiety of a tumor-reactive monoclonal antibody. Naptumomab estafenatox targets the 5T4 tumor antigen, a 72-kDa oncofetal trophoblast protein expressed on many carcinomas, including renal cell carcinoma. The therapeutic effect is associated with activation of SAg-binding T cells. The SAg-binding T lymphocytes expand, differentiate to effector cells, and infiltrate the tumor. The therapeutic efficacy is most likely related to the dual mechanism of tumor cell killing: (1) direct lysis by cytotoxic T lymphocytes of tumor cells expressing the antigen recognized by the antibody moiety of the fusion protein and (2) secretion of cytokines eliminating antigen-negative tumor cell variants. Naptumomab estafenatox has been clinically tested in a range of solid tumors with focus on renal cell carcinoma. This review looks at the clinical experience with the new immunotoxin and its potential.
Collapse
Affiliation(s)
- Tim Eisen
- Cambridge University Health Partners, Addenbrooke's Hospital, Cambridge, UK,
| | | | | | | |
Collapse
|
31
|
Understanding and exploiting 5T4 oncofoetal glycoprotein expression. Semin Cancer Biol 2014; 29:13-20. [PMID: 25066861 DOI: 10.1016/j.semcancer.2014.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/17/2014] [Indexed: 01/14/2023]
Abstract
Oncofoetal antigens are present during foetal development with generally limited expression in the adult but are upregulated in cancer. These molecules can sometimes be used to diagnose or follow treatment of tumours or as a target for different immunotherapies. The 5T4 oncofoetal glycoprotein was identified by searching for shared surface molecules of human trophoblast and cancer cells with the rationale that they may function to allow survival of the foetus as a semi-allograft in the mother or a tumour in its host, potentially influencing growth, invasion or altered immune surveillance of the host. 5T4 tumour selective expression has stimulated the development of 5T4 vaccine, 5T4 antibody targeted-superantigen and 5T4 antibody-drug therapies through preclinical and into clinical studies. It is now apparent that 5T4 expression is a marker of the use (or not) of several cellular pathways relevant to tumour growth and spread. Thus 5T4 expression is mechanistically associated with the directional movement of cells through epithelial mesenchymal transition, facilitation of CXCL12/CXCR4 chemotaxis, blocking of canonical Wnt/beta-catenin while favouring non-canonical pathway signalling. These processes are highly regulated in development and in normal adult tissues but can contribute to the spread of cancer cells. Understanding the differential impact of these pathways marked by 5T4 can potentially improve existing, or aid development of novel cancer treatment strategies.
Collapse
|
32
|
Scurr M, Bloom A, Pembroke T, Srinivasan R, Brown C, Smart K, Bridgeman H, Davies M, Hargest R, Phillips S, Christian A, Hockey T, Gallimore A, Godkin A. Escalating regulation of 5T4-specific IFN-γ + CD4 + T cells distinguishes colorectal cancer patients from healthy controls and provides a target for in vivo therapy. Cancer Immunol Res 2013; 1. [PMID: 24409450 DOI: 10.1158/2326-6066.cir-13-0035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The relationship between the adaptive CD4+ T cell response and human cancer is unclear. The oncofetal antigen 5T4 is expressed on many human carcinomas, including colorectal cancer (CRC) cells, but has limited expression on normal tissues. We previously identified anti-5T4 CD4+ T cells in a proportion of CRC patients, and we extended this study to examine whether the quality or quantity of the T cell response reflects tumor stage. An overlapping peptide library spanning 5T4 was used as a target to enumerate cognate IFN-γ+CD4+ T-cells (measured as spot forming cells [SFC]/105 cultured T cells) in peripheral blood-derived lymphocytes following a 12-day in vitro culture period comparing patients pre-operatively (n = 27) to healthy controls (n = 17). Robust 5T4-specific T cell responses were present in 100% of healthy donors. There was a steady loss of T cell responses with advancing tumors with a significant negative correlation from stage I to III (P = 0.008). The predictability of the decline meant < 200 SFC/105 was only found in subjects with stage III CRC. The mechanism of loss of T cell response is independent of HLA-DR type or patient age, but does correspond to increases in Foxp3+ regulatory T cells (Tregs). Using low-dose cyclophosphamide to reduce the proportion of Tregs in vivo resulted in increased anti-5T4 T cell responses in CRC patients. The selective loss of 5T4-specific IFN-γ+CD4+ T cell responses implies a link between tumor stage and antitumor Th1 effector function; depleting Tregs can enhance such responses.
Collapse
Affiliation(s)
- Martin Scurr
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Anja Bloom
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Tom Pembroke
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Rohit Srinivasan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Clare Brown
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Kathryn Smart
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Hayley Bridgeman
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Mike Davies
- Department of Surgery, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Rachel Hargest
- Department of Surgery, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Simon Phillips
- Department of Surgery, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Adam Christian
- Department of Histopathology, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Tom Hockey
- Department of Histopathology, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Awen Gallimore
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Andrew Godkin
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
33
|
Cruciat CM, Niehrs C. Secreted and transmembrane wnt inhibitors and activators. Cold Spring Harb Perspect Biol 2013; 5:a015081. [PMID: 23085770 DOI: 10.1101/cshperspect.a015081] [Citation(s) in RCA: 494] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signaling by the Wnt family of secreted glycoproteins plays important roles in embryonic development and adult homeostasis. Wnt signaling is modulated by a number of evolutionarily conserved inhibitors and activators. Wnt inhibitors belong to small protein families, including sFRP, Dkk, WIF, Wise/SOST, Cerberus, IGFBP, Shisa, Waif1, APCDD1, and Tiki1. Their common feature is to antagonize Wnt signaling by preventing ligand-receptor interactions or Wnt receptor maturation. Conversely, the Wnt activators, R-spondin and Norrin, promote Wnt signaling by binding to Wnt receptors or releasing a Wnt-inhibitory step. With few exceptions, these antagonists and agonists are not pure Wnt modulators, but also affect additional signaling pathways, such as TGF-β and FGF signaling. Here we discuss their interactions with Wnt ligands and Wnt receptors, their role in developmental processes, as well as their implication in disease.
Collapse
Affiliation(s)
- Cristina-Maria Cruciat
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, DKFZ, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | |
Collapse
|
34
|
Abstract
Many tissues of the body cannot only repair themselves, but also self-renew, a property mainly due to stem cells and the various mechanisms that regulate their behavior. Stem cell biology is a relatively new field. While advances are slowly being realized, stem cells possess huge potential to ameliorate disease and counteract the aging process, causing its speculation as the next panacea. Amidst public pressure to advance rapidly to clinical trials, there is a need to understand the biology of stem cells and to support basic research programs. Without a proper comprehension of how cells and tissues are maintained during the adult life span, clinical trials are bound to fail. This review will cover the basic biology of stem cells, the various types of stem cells, their potential function, and the advantages and disadvantages to their use in medicine. We will next cover the role of G protein-coupled receptors in the regulation of stem cells and their potential in future clinical applications.
Collapse
Affiliation(s)
- VAN A. DOZE
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA (V.A.D.), and Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA (D.M.P.)
| | - DIANNE M. PEREZ
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA (V.A.D.), and Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA (D.M.P.)
| |
Collapse
|
35
|
Sapra P, Damelin M, DiJoseph J, Marquette K, Geles KG, Golas J, Dougher M, Narayanan B, Giannakou A, Khandke K, Dushin R, Ernstoff E, Lucas J, Leal M, Hu G, O'Donnell CJ, Tchistiakova L, Abraham RT, Gerber HP. Long-term Tumor Regression Induced by an Antibody–Drug Conjugate That Targets 5T4, an Oncofetal Antigen Expressed on Tumor-Initiating Cells. Mol Cancer Ther 2012; 12:38-47. [DOI: 10.1158/1535-7163.mct-12-0603] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
McGinn OJ, Marinov G, Sawan S, Stern PL. CXCL12 receptor preference, signal transduction, biological response and the expression of 5T4 oncofoetal glycoprotein. J Cell Sci 2012; 125:5467-78. [PMID: 22956548 DOI: 10.1242/jcs.109488] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
CXCL12 is a pleiotropic chemokine capable of eliciting multiple signal transduction cascades and functions, via interaction with either CXCR4 or CXCR7. Factors that determine CXCL12 receptor preference, intracellular signalling route and biological response are poorly understood but are of central importance in the context of therapeutic intervention of the CXCL12 axis in multiple disease states. We have recently demonstrated that 5T4 oncofoetal glycoprotein facilitates functional CXCR4 expression leading to CXCL12 mediated chemotaxis in mouse embryonic cells. Using wild type (WT) and 5T4 knockout (5T4KO) murine embryonic fibroblasts (MEFs), we now show that CXCL12 binding to CXCR4 activates both the ERK and AKT pathways within minutes, but while these pathways are intact, they are non-functional in 5T4KO cells treated with CXCL12. Importantly, in the absence of 5T4 expression, CXCR7 is upregulated and becomes the predominant receptor for CXCL12, activating a distinct signal transduction pathway with slower kinetics involving transactivation of the epidermal growth factor receptor (EGFR), eliciting proliferation rather than chemotaxis. Thus the surface expression of 5T4 marks the use of the CXCR4 rather than the CXCR7 receptor, with distinct consequences for CXCL12 exposure, relevant to the spread and growth of a tumour. Consistent with this hypothesis, we have identified human small cell lung carcinoma cells with similar 5T4/CXCR7 reciprocity that is predictive of biological response to CXCL12 and determined that 5T4 expression is required for functional chemotaxis in these cells.
Collapse
Affiliation(s)
- Owen J McGinn
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester M13 9PT, UK
| | | | | | | |
Collapse
|
37
|
Amato RJ, Stepankiw M. Evaluation of MVA-5T4 as a novel immunotherapeutic vaccine in colorectal, renal and prostate cancer. Future Oncol 2012; 8:231-7. [PMID: 22409460 DOI: 10.2217/fon.12.7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This paper reviews the development of the combination of modified vaccinia Ankara (MVA) to deliver the tumor-associated antigen 5T4 as a novel immunotherapeutic vaccine. The oncofetal antigen 5T4 is highly expressed in 80% of breast, kidney, colorectal, prostate and ovarian carcinomas, making it an ideal antigen for vaccine therapy. To date, more than 3000 doses of MVA-5T4 have been administered to colorectal, renal and prostate cancer patients, with rare occurrences of grade 3 or 4 vaccination-related adverse events being observed. Studies have demonstrated that MVA-5T4 is safe and highly immunogenic, both as monotherapy and in combination with other standard of care therapies. Although an immune response has been observed, antitumor activity has been modest or absent in clinical trials. A Phase III trial resulted in the development of an immune response surrogate that is to be applied to all future MVA-5T4 clinical trials. With minimal side effects and the ability to produce a strong immunogenic response, MVA-5T4 is a viable addition to the cancer therapy arsenal.
Collapse
Affiliation(s)
- Robert J Amato
- The University of Texas Health Science Center at Houston, 6410 Fannin St Suite 830, Houston, TX 77030, USA.
| | | |
Collapse
|
38
|
Castro FV, Al-Muftah M, Mulryan K, Jiang HR, Drijfhout JW, Ali S, Rutkowski AJ, Kalaitsidou M, Gilham DE, Stern PL. Regulation of autologous immunity to the mouse 5T4 oncofoetal antigen: implications for immunotherapy. Cancer Immunol Immunother 2012; 61:1005-18. [PMID: 22127365 PMCID: PMC11029011 DOI: 10.1007/s00262-011-1167-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/16/2011] [Indexed: 02/06/2023]
Abstract
Effective vaccination against tumour-associated antigens (TAA) such as the 5T4 oncofoetal glycoprotein may be limited by the nature of the T cell repertoire and the influence of immunomodulatory factors in particular T regulatory cells (Treg). Here, we identified mouse 5T4-specific T cell epitopes using a 5T4 knock out (5T4KO) mouse and evaluated corresponding wild-type (WT) responses as a model to refine and improve immunogenicity. We have shown that 5T4KO mice vaccinated by replication defective adenovirus encoding mouse 5T4 (Adm5T4) generate potent 5T4-specific IFN-γ CD8 and CD4 T cell responses which mediate significant protection against 5T4 positive tumour challenge. 5T4KO CD8 but not CD4 primed T cells also produced IL-17. By contrast, Adm5T4-immunized WT mice showed no tumour protection consistent with only low avidity CD8 IFN-γ, no IL-17 T cell responses and no detectable CD4 T cell effectors producing IFN-γ or IL-17. Treatment with anti-folate receptor 4 (FR4) antibody significantly reduced the frequency of Tregs in WT mice and enhanced 5T4-specific IFN-γ but reduced IL-10 T cell responses but did not reveal IL-17-producing effectors. This altered balance of effectors by treatment with FR4 antibody after Adm5T4 vaccination provided modest protection against autologous B16m5T4 melanoma challenge. The efficacy of 5T4 and some other TAA vaccines may be limited by the combination of TAA-specific T regs, the deletion and/or alternative differentiation of CD4 T cells as well as the absence of distinct subsets of CD8 T cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Epitopes, T-Lymphocyte/immunology
- Immunotherapy, Active/methods
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Receptors, Cell Surface/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Fernanda V. Castro
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
| | - Mariam Al-Muftah
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
- Clinical and Experimental Immunotherapy, Medical Oncology, School of Cancer and Enabling Sciences, University of Manchester, Manchester Academic Healthcare Science Centre, Manchester, UK
| | - Kate Mulryan
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
| | - Hui-Rong Jiang
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Jan-Wouter Drijfhout
- Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Sumia Ali
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
| | - Andrzej J. Rutkowski
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
| | - Milena Kalaitsidou
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
| | - David E. Gilham
- Clinical and Experimental Immunotherapy, Medical Oncology, School of Cancer and Enabling Sciences, University of Manchester, Manchester Academic Healthcare Science Centre, Manchester, UK
| | - Peter L. Stern
- Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester, M20 4BX UK
| |
Collapse
|
39
|
Al-Taei S, Salimu J, Lester JF, Linnane S, Goonewardena M, Harrop R, Mason MD, Tabi Z. Overexpression and potential targeting of the oncofoetal antigen 5T4 in malignant pleural mesothelioma. Lung Cancer 2012; 77:312-8. [PMID: 22498111 DOI: 10.1016/j.lungcan.2012.03.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/02/2012] [Accepted: 03/14/2012] [Indexed: 01/18/2023]
Abstract
Malignant pleural mesothelioma (MPM) is resistant to conventional treatments. Novel, targeted treatments are hampered by the relative lack of MPM-associated tumour antigens. The aim of this study was to evaluate the level of expression and the relevance of 5T4 as a tumour-associated antigen in MPM. 5T4 expression was assessed by Western blotting, flow cytometry, immuno-cytochemistry and -histochemistry in 11 mesothelioma cell lines, 21 tumour biopsies, and ex vivo tumour cells obtained from the pleural fluid (PF) of 10 patients. 5T4 antibody levels were also determined in the plasma of patients and healthy donors. The susceptibility of MPM cells to 5T4-specific T-cell-mediated killing was determined using an HLA-A2(+), CD8(+) T-cell line, developed against the 5T4(17-25) peptide. We report here that cell surface 5T4 expression was detected in all mesothelioma cell lines and PF cell samples. Mesothelin and CD200, a suggested mesothelioma marker, were co-expressed with 5T4 on tumour cells in PF. Immunohistochemistry confirmed overexpression of 5T4, similar to mesothelin, on tumour cells but not on reactive stroma in all tissue sections tested. Median 5T4 antibody levels were 46% higher in patient than in healthy donor plasma, indicating immune recognition. Importantly, 5T4-specific CD8(+) T-cells were able to kill four out of six HLA-A2(+) MPM cell lines but not an HLA-A2(-) cell line, demonstrating immune recognition of MPM-associated 5T4 antigen at the effector T-cell level. We conclude that 5T4 is a potential new antigen for targeted therapies such as immunotherapy in MPM, as it is overexpressed on mesothelioma cells and recognised by 5T4-specific cytotoxic T-cells. Our findings have been translated into a Phase II clinical trial applying 5T4-targeted therapies in MPM patients.
Collapse
Affiliation(s)
- Saly Al-Taei
- Department of Oncology, School of Medicine, Cardiff University, Velindre Cancer Centre, Velindre Road, Whitchurch, Cardiff CF14 2TL, UK
| | | | | | | | | | | | | | | |
Collapse
|
40
|
5T4 glycoprotein regulates the sensory input-dependent development of a specific subtype of newborn interneurons in the mouse olfactory bulb. J Neurosci 2012; 32:2217-26. [PMID: 22323733 DOI: 10.1523/jneurosci.5907-11.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Sensory input has been shown to regulate development in a variety of species and in various structures, including the retina, cortex, and olfactory bulb (OB). Within the mammalian OB specifically, the development of dendrites in mitral/tufted cells is well known to be odor-evoked activity dependent. However, little is known about the developmental role of sensory input in the other major OB population of the GABAgenic interneurons, such as granule cells and periglomerular cells. Here, we identified, with DNA microarray and in situ hybridization screenings, a trophoblast glycoprotein gene, 5T4, whose expression in a specific subtype of OB interneurons is dependent on sensory input. 5T4 is a type I membrane protein, whose extracellular domain contains seven leucine-rich repeats (LRR) flanked by characteristic LRR-N-flanking and C-flanking regions, and a cytoplasmic domain. 5T4 overexpression in the newborn OB interneurons facilitated their dendritic arborization even under the sensory input-deprived condition. By contrast, both 5T4 knockdown with RNAi and 5T4 knockout with mice resulted in a significant reduction in the dendritic arborization of 5T4(+) granule cells. Further, we identified the amino acid sequence in the 5T4 cytoplasmic domain that is necessary and sufficient for the sensory input-dependent dendritic shaping of specific neuronal subtypes in the OB. Thus, these results demonstrate that 5T4 glycoprotein contributes in the regulation of activity-dependent dendritic development of interneurons and the formation of functional neural circuitry in the OB.
Collapse
|
41
|
Castro FV, McGinn OJ, Krishnan S, Marinov G, Li J, Rutkowski AJ, Elkord E, Burt DJ, Holland M, Vaghjiani R, Gallego A, Saha V, Stern PL. 5T4 oncofetal antigen is expressed in high risk of relapse childhood pre-B acute lymphoblastic leukemia and is associated with a more invasive and chemotactic phenotype. Leukemia 2012; 26:1487-98. [PMID: 22266911 PMCID: PMC3378689 DOI: 10.1038/leu.2012.18] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Although the overall prognosis in childhood acute lymphoblastic leukemia (ALL) is good, outcome after relapse is poor. Recurrence is frequently characterized by the occurrence of disease at extramedullary sites, such as the central nervous system and testes. Subpopulations of blasts able to migrate to such areas may have a survival advantage and give rise to disease recurrence. Gene expression profiling of 85 diagnostic pre-B-ALL bone marrow samples revealed higher 5T4 oncofetal antigen transcript levels in cytogenetic high-risk subgroups of patients (P<0.001). Flow cytometric analysis determined that bone marrow from relapse patients have a significantly higher percentage of 5T4-positive leukemic blasts than healthy donors (P=0.005). The high-risk Sup-B15 pre-B-ALL line showed heterogeneity in 5T4 expression, and the derived, 5T4(+) (Sup5T4) and 5T4(-) (Sup) subline cells, displayed differential spread to the omentum and ovaries following intraperitoneal inoculation of immunocompromised mice. Consistent with this, Sup5T4 compared with Sup cells show increased invasion in vitro concordant with increased LFA-1 and VLA-4 integrin expression, adhesion to extracellular matrix and secretion of matrix metalloproteases (MMP-2/-9). We also show that 5T4-positive Sup-B15 cells are susceptible to 5T4-specific superantigen antibody-dependent cellular toxicity providing support for targeted immunotherapy in high-risk pre-B-ALL.
Collapse
Affiliation(s)
- F V Castro
- Cancer Research UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kagermeier-Schenk B, Wehner D, Ozhan-Kizil G, Yamamoto H, Li J, Kirchner K, Hoffmann C, Stern P, Kikuchi A, Schambony A, Weidinger G. Waif1/5T4 inhibits Wnt/β-catenin signaling and activates noncanonical Wnt pathways by modifying LRP6 subcellular localization. Dev Cell 2011; 21:1129-43. [PMID: 22100263 DOI: 10.1016/j.devcel.2011.10.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 09/09/2011] [Accepted: 10/20/2011] [Indexed: 02/03/2023]
Abstract
Wnt proteins can activate distinct signaling pathways, but little is known about the mechanisms regulating pathway selection. Here we show that the metastasis-associated transmembrane protein Wnt-activated inhibitory factor 1 (Waif1/5T4) interferes with Wnt/β-catenin signaling and concomitantly activates noncanonical Wnt pathways. Waif1 inhibits β-catenin signaling in zebrafish and Xenopus embryos as well as in mammalian cells, and zebrafish waif1a acts as a direct feedback inhibitor of wnt8-mediated mesoderm and neuroectoderm patterning during zebrafish gastrulation. Waif1a binds to the Wnt coreceptor LRP6 and inhibits Wnt-induced LRP6 internalization into endocytic vesicles, a process that is required for pathway activation. Thus, Waif1a modifies Wnt/β-catenin signaling by regulating LRP6 subcellular localization. In addition, Waif1a enhances β-catenin-independent Wnt signaling in zebrafish embryos and Xenopus explants by promoting a noncanonical function of Dickkopf1. These results suggest that Waif1 modulates pathway selection in Wnt-receiving cells.
Collapse
Affiliation(s)
- Birgit Kagermeier-Schenk
- Biotechnology Center and Center for Regenerative Therapies, Technische Universität Dresden, Tatzberg 47, 01307 Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Damelin M, Geles KG, Follettie MT, Yuan P, Baxter M, Golas J, DiJoseph JF, Karnoub M, Huang S, Diesl V, Behrens C, Choe SE, Rios C, Gruzas J, Sridharan L, Dougher M, Kunz A, Hamann PR, Evans D, Armellino D, Khandke K, Marquette K, Tchistiakova L, Boghaert ER, Abraham RT, Wistuba II, Zhou BBS. Delineation of a cellular hierarchy in lung cancer reveals an oncofetal antigen expressed on tumor-initiating cells. Cancer Res 2011; 71:4236-46. [PMID: 21540235 DOI: 10.1158/0008-5472.can-10-3919] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Poorly differentiated tumors in non-small cell lung cancer (NSCLC) have been associated with shorter patient survival and shorter time to recurrence following treatment. Here, we integrate multiple experimental models with clinicopathologic analysis of patient tumors to delineate a cellular hierarchy in NSCLC. We show that the oncofetal protein 5T4 is expressed on tumor-initiating cells and associated with worse clinical outcome in NSCLC. Coexpression of 5T4 and factors involved in the epithelial-to-mesenchymal transition were observed in undifferentiated but not in differentiated tumor cells. Despite heterogeneous expression of 5T4 in NSCLC patient-derived xenografts, treatment with an anti-5T4 antibody-drug conjugate resulted in complete and sustained tumor regression. Thus, the aggressive growth of heterogeneous solid tumors can be blocked by therapeutic agents that target a subpopulation of cells near the top of the cellular hierarchy.
Collapse
Affiliation(s)
- Marc Damelin
- Oncology Research Unit, Pfizer Inc., Pearl River, New York, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Neutralizing anti-gH antibody of Varicella-zoster virus modulates distribution of gH and induces gene regulation, mimicking latency. J Virol 2011; 85:8172-80. [PMID: 21632752 DOI: 10.1128/jvi.00435-11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The anti-glycoprotein H (gH) monoclonal antibody (anti-gH-MAb) that neutralizes varicella-zoster virus (VZV) inhibited cell-to-cell infection, resulting in a single infected cell without apoptosis or necrosis, and the number of infectious cells in cultures treated with anti-gH-MAb declined to undetectable levels in 7 to 10 days. Anti-gH-MAb modulated the wide cytoplasmic distribution of gH colocalized with glycoprotein E (gE) to the cytoplasmic compartment with endoplasmic reticulum (ER) and Golgi markers near the nucleus, while gE retained its cytoplasmic distribution. Thus, the disintegrated distribution of gH and gE caused the loss of cellular infectivity. After 4 weeks of treatment with anti-gH-MAb, no infectious virus was recovered, even after cultivation without anti-gH-MAb for another 8 weeks or various other treatments. Cells were infected with Oka varicella vaccine expressing hepatitis B surface antigen (ROka) and treated with anti-gH-MAb for 4 weeks, and ROka was recovered from the quiescently infected cells by superinfection with the parent Oka vaccine. Among the genes 21, 29, 62, 63, and 66, transcripts of gene 63 were the most frequently detected, and products from the genes 63 and 62, but not gE, were detected mainly in the cytoplasm of quiescently infected cells, in contrast to their nuclear localization in lytically infected cells. The patterns of transcripts and products from the quiescently infected cells were similar to those of latent VZV in human ganglia. Thus, anti-gH-MAb treatment resulted in the antigenic modulation and dormancy of infectivity of VZV. Antigenic modulation by anti-gH-MAb illuminates a new aspect in pathogenesis in VZV infection and the gene regulation of VZV during latency in human ganglia.
Collapse
|
45
|
Kobayashi NR, Hawes SM, Crook JM, Pébay A. G-protein coupled receptors in stem cell self-renewal and differentiation. Stem Cell Rev Rep 2010; 6:351-66. [PMID: 20625855 DOI: 10.1007/s12015-010-9167-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cells have great potential for understanding early development, treating human disease, tissue trauma and early phase drug discovery. The factors that control the regulation of stem cell survival, proliferation, migration and differentiation are still emerging. Some evidence now exists demonstrating the potent effects of various G-protein coupled receptor (GPCR) ligands on the biology of stem cells. This review aims to give an overview of the current knowledge of the regulation of embryonic and somatic stem cell maintenance and differentiation by GPCR ligands.
Collapse
|