Unstable B7-H4 cell surface expression and T-cell redirection as a means of cancer therapy

  • Authors:
    • Akira Iizuka
    • Ryota Kondou
    • Chizu Nonomura
    • Tadashi Ashizawa
    • Keiichi Ohshima
    • Masatoshi Kusuhara
    • Mitsuhiro Isaka
    • Yasuhisa Ohde
    • Ken Yamaguchi
    • Yasuto Akiyama
  • View Affiliations

  • Published online on: September 12, 2016     https://doi.org/10.3892/or.2016.5084
  • Pages: 2625-2632
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor immune regulation has been demonstrated in clinical studies using antibodies targeted to the B7/CD28 family. B7 homolog 4 (B7-H4) negatively regulates immune responses and is overexpressed in many types of human cancer, indicating that B7-H4 may be a potential target of cancer therapy. B7-H4 expression is affected by the microenvironment, and its presence has been reported in cancer tissues and immune cells. We found an upregulation of B7-H4 expression using comprehensive whole exome sequencing and gene expression profiling (project HOPE) launched by the Shizuoka Cancer Center based on tumor tissue samples from 1,058 cancer patients. We were successful in producing monoclonal antibodies for B7-H4 and demonstrated B7-H4 dimerization and rapid cell surface disappearance by antibody cross-linking in breast cancer cells, even under typical conditions. These observations may explain why antibody-dependent cellular cytotoxicity (ADCC) did not function in vivo on the B7-H4-expressing tumor cells. Unstable cell surface antigens are not suitable as targets for ADCC, and we therefore performed an indirect ADCC-redirecting T-cell cytotoxicity assay to study B7-H4 using polyclonal anti-mouse IgG antibody-mediated linking. Our results showed the possibility of targeting the B7-H4 molecule as a means of treating cancer.

Introduction

T-cell dependent tumor immune responses have been demonstrated in clinical studies and the CD28 family of receptors and the B7 family of ligands are thought to be most attractive as T-cell regulatory targets for cancer immunotherapy or for the control of immunological disorders (14). B7 homolog 4 (B7-H4) negatively regulates T-cell proliferation and cytokine production (5,6). B7-H4 mRNA is expressed widely in multiple tissues to a low degree (6,7), but its receptor on activated lymphocytes has yet to be identified (8,9).

B7-H4 is overexpressed in many human cancers (1013), and B7-H4 levels in patient sera are upregulated in ovarian cancer and renal cell carcinoma (RCC) (14,15). However, low levels of B7-H4 can also be detected in healthy human sera. Endothelial cells of the RCC tumor vasculature express B7-H4, whereas normal renal vessels and tissues exhibit little to no expression. Notably, patients with RCC expressing both B7-H1 and B7-H4 have a poor prognosis compared to patients presenting with single expression alone or patients with no expression at all (12). By contrast, high expression of B7-H4 in breast cancer is correlated with an improved recurrence-free survival (16).

Ovarian cancer patients frequently express the B7-H4 ligand in the cytoplasm and on membranes but not on the cell surface (17,18), and the expression level is inversely correlated with patient survival (19). However, B7-H4-expressing infiltrating macrophages, but not tumor cells, suppress tumor-associated antigen (TAA)-specific T-cell immunity (18). Tumor cell-derived B7-H4 could function in immunosuppression even though the physiological implications are not apparent.

IL-6 and IL-10 induce B7-H4 expression on tumor-associated macrophages (TAMs) and on other immune cells involved in Treg development (18,19), but IL-6 and IL-10 receptors rarely exist on normal tissue cells (20) and non-hematopoietic malignanct cell lines (21), which indicates that B7-H4 expression in tumor cells is regulated in a different manner than in immune cells. By contrast, IFN-γ induces B7-H4 expression on mouse embryonic fibroblasts (16), and its receptor is widely expressed on both normal and tumor cells. This indicates that IFN-γ is prossibly a B7-H4 inducer in tumors. Consequently, ovarian cancer rapidly loses B7-H4 expression after a few days in vitro, which suggests that the microenvironment can influence B7-H4 expression (22).

B7-H4 is a candidate immunoregulatory target and may also be a direct therapeutic target against tumors. B7-H4 knockdown by morpholino oligos improved TAA-specific T-cell immunity (18) and knockout protected mice from experimental lung metastasis (23). However, antibody-dependent cellular cytotoxicity (ADCC) or tumor growth suppression was not reported except during B7-H4 forced expression (11). Notably, Lee et al reported that B7-H4 is not detected on immune cells of either humans or mice by flow cytometry before or even after stimulation, and this observation is inconsistent with previous studies (24).

In clinical studies targeting immune checkpoint molecules of the B7/CD28 families, a B7-H1 (PD-L1)-targeting inhibitory antibody has shown potent antitumor effects in advanced melanoma (3), although B7-H4-targeting clinical trials have not yet been performed.

In this study, we performed comprehensive whole exome sequencing and gene expression profiling based on 1,058 cancer patient-derived tumor tissues and consequently found high B7-H4 expression in non-small cell lung cancer and breast cancer patients. Furthermore, we characterized unstable expression of the B7-H4 molecule in cancer cells using antibodies generated in house. Finally, we discuss how to use anti-B7-H4 antibodies for cancer immunotherapy.

Materials and methods

Patient materials

This clinical research project using comprehensive whole exome sequencing and gene expression profiling of various tumor tissues, called the High-tech Omics-based Patient Evaluation (HOPE) for Cancer Therapy, has been conducted in accordance with the ʻEthical Guidelines for Human Genome and Genetic Analysis Researchʼ, revised in 2013. Informed consents were obtained from all patients participating in the HOPE project, which was approved by the Institutional Review Board of the Shizuoka Cancer Center (SCC), Japan. Tumor tissues, along with surrounding normal tissues, were dissected from surgical specimens by trained pathologists.

Animal experiments

BALB/cA mice and nude mice (BALB/cA-nu/nu) were obtained from Nippon Clea (Tokyo, Japan). All animals were cared for and used humanely according to the Guidelines for the Welfare and Use of Animals in Cancer Research (25). All procedures were approved by the Animal Care and Use Committee of the Shizuoka Cancer Center (SCC) Research Institute.

Gene expression analysis of the B7 family of genes

Total RNA was extracted from ~10 mg of tissue samples using the miRNeasy Mini Kit (Qiagen) according to the manufacturer's instructions. Following assessment using an Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA), total RNA with an RNA integrity number (RIN) of six or higher was used for DNA microarray analysis. Gene expression analysis was performed using SurePrint G3 Human GE 8×60K v2.0 arrays (Agilent Technologies) according to the manufacturer's instructions. Signal data analysis was carried out using GeneSpring version 13.1.1 software (Agilent Technologies). The ratio of tumor tissue vs. surrounding non-cancerous tissue was calculated from the normalized values.

Generation of anti-B7-H4 monoclonal antibodies

The human B7-H4 isoform 1 extracellular domain was constructed with a 6X histidine tag in a pcDNA3.3 expression vector, and the B7-H4 extracellular soluble free form was harvested from Expi293F™ cell (Life Technologies Corporation) culture supernatant, affinity-purified, and used for experiments. B7-H1 and B7-DC were produced in the same way for use as negative controls. After the immunization of BALB/cA mice, an antibody secreting hybridoma was generated by a common method using the mouse myeloma cell line P3X63Ag8.653 (American Type Culture Collection; ATCC, Manassas, VA, USA).

ELISA

Specificity was validated by sandwich ELISA and fluorescent staining of B7-H4-transfected HEK293 cells. Briefly, purified newly-generated antibodies were immobilized on a 96-well microplate Nunc Immobilizer Amino Surface (Thermo Fisher Scientific, Inc.) prior to the addition of 3% bovine serum albumin for overnight blocking and 10 µg/ml B7-H4 (soluble form) or controls. After being washed, 2 µg/ml biotinylated antibody (Ab) clone #25 was added and detected by HRP-conjugated streptavidin (Thermo Fisher Scientific, Inc.).

Flow cytometry

HEK293 cells that transformed with the full B7-H4 sequence or cancer cell lines were incubated with 8 µg/ml Ab clones and later incubated with 4 µg/ml PE-labeled polyclonal anti-mouse Ig Ab (BD Biosciences) on ice. Fluorescence intensity was determined by the flow cytometer FACSCanto (BD Biosciences).

Affinity kinetics determination of anti-B7-H4 antibodies

Surface plasmon resonance (SPR) analysis was performed on a Biacore ×100 (GE Healthcare) in order to determine the kinetics of the anti-B7-H4 Ab and free form of B7-H4. All reagents and sensor chips were purchased from GE Healthcare. Immobilization of anti-B7-H4 IgG antibodies on the CM5 sensor chip was performed at pH 5.0, and the Ab binding was targeted to 1000 response units (RU). To regenerate the sensor chip, 10 mM glycine-HCl pH 1.7 was used. The binding kinetics were monitored by injecting multiple concentrations of B7-H4 in HBS buffer (10 mM HEPES pH 7.4, containing 0.15 M NaCl, 3 mM EDTA, and 0.05% Tween-20) at a flow rate of 30 µl/min at 25°C. The dissociation phase was also monitored by HBS buffer flow. The binding kinetic parameters were calculated by Biacore ×100 evaluation software.

Western blot analysis

Breast cancer cell line lysates harvested with Laemmli sample buffer, and over-confluent culture supernatants were used for western blot analysis. Cell lysates and supernatants were electrophoresed through a SDS-PAGE gradient gel and transferred to a PVDF membrane. After overnight blocking with 5% skim milk and washing with 0.05% Tween-20-PBS (T-PBS), the membrane was incubated for 1 h at room temperature (RT) with rabbit anti-human B7-H4 Ab (clone, EP1165). After being washed with T-PBS, the membrane was incubated with a secondary Ab, HRP-conjugated donkey anti-rabbit IgG polyclonal Ab (GE Healthcare) diluted 1:10,000. ECL Plus (GE Healthcare) was used for detection according to the manufacturer's instructions.

Immunohistochemistry

Female nude mice (BALB/cA-nu/nu, 5–6 weeks old) were transplanted subcutaneously with human breast cancer MDA-MB-468 or lung cancer NCI-H2170 cells. Formalin-fixed paraffin-embedded (FFPE) cancer tissue blocks were made. To evaluate B7-H4 expression, sections from the cancers were immunostained with anti-B7-H4 Ab (clone #25) or mouse IgG1 isotype control (BD Pharmingen) and later stained with hematoxylin.

Tumor growth inhibition assay

MDA-MB-468 cells (1×107) were inoculated into the mammary fat pad of BALB/cA-nu/nu mice. Antibody clone #25, at doses of 2 or 10 mg/kg, was administered by intraperitoneal injection twice a week from day 9 to day 27 after tumor inoculation. To evaluate the antitumor activity against inoculated tumors, tumor volume was calculated based on the National Cancer Institute formula as follows: Tumor volume (mm3) = length (mm) × [width (mm)]2 × 1/2.

Indirect ADCC experiment

Briefly, effector cells were isolated from healthy volunteer peripheral blood by Ficoll-Paque PLUS (GE Healthcare Life Sciences, Buckinghamshire, UK) and then enriched by depletion of CD14+ or CD19+ cells using MicroBeads and autoMACS (Miltenyi Biotec K.K., Tokyo, Japan). The effector cells were incubated on ice for 10 min with 0.2 mg/ml (high concentration) anti-CD3 Ab (OKT3) diluted with staining buffer (0.5% BSA, 0.1% azide, 1 mM glucose-PBS), followed by washing. Cells were then incubated with 0.5 mg/ml polyclonal rabbit anti-mouse immunoglobulins (DakoCytomation, Glostrup, Denmark) and after being washed, they were incubated with 0.5 mg/ml of each anti-B7-H4 antibody. The effector cells were washed and resuspended to a concentration of ~107 cells/ml in 10% FBS-RPMI-1640 and co-cultured with non-radioactive reagent-labeled MDA-MB-468 target cells in a 96-well round-bottom plate at 37°C for 3 h. Target cell lysis was performed with DELFIA® EuTDA Cytotoxicity Assay Reagents and measured using a Wallac 1420 ARVO SX multilabel counter (PerkinElmer, Waltham, MA, USA). The percentage of specific lysis was determined by the following formula: Percentage of specific lysis = (experimental release − spontaneous release)/(maximal release − spontaneous release) × 100.

Statistical analysis

Statistically differences were analyzed using the Student's paired two-tailed t-test. Values of P<0.01 were considered to indicate statistically significant differences.

Results

B7-H4 expression in cancer tissues and cell lines

We found that B7-H4 expression was upregulated in some types of cancer when compared to expression in non-cancerous normal tissues (Fig. 1). B7-H4 expression was upregulated in breast, lung, and gynecological cancers and downregulated in skin and urinary tract cancers. Expression of other genes from the B7 family was not apparently altered. B7-H4 expression was detected in breast and ovarian cancer cell lines by RT-PCR in accordance with the previous study (data not shown), but the soluble free form of B7-H4 in the culture medium was not, as determined by ELISA. The splice variants of B7-H4 that were detected corresponded mainly to isoforms 1, 2, and 4 and partially to isoform 3 which has a different sequence.

Production and characterization of anti-B7-H4 monoclonal antibodies

We generated 19 hybridoma clones of anti-human B7-H4 isoform 1 for detection of the natural form, and purified eight antibody clones which could be cultured in vitro under serum-free conditions. The antibody clones bound B7-H4 but not B7 homologs PD-L1 or PD-L2 (B7-DC) (Fig. 2A), and the staining of HEK293 cells transfected with the full B7-H4 isoform 1 sequence showed two different types of staining, strong (#18, #25) and weak (#113, #121, #209), by flow cytometry. All mAb clones were confirmed to have different DNA sequences (Fig. 2B).

B7-H4 forms homodimers

Sandwich ELISAs using antibody clone #25 as both the capture and detector antibody detected the free form of B7-H4; ELISA using clone #18 as the capture Ab showed a similar curve. These two capture Abs had lower sensitivities than the other clones, which contrasted to staining of HEK293 transfectants (Figs. 3A and 2B). B7-H4 is a monovalent molecule, and thus these observations were indicative of B7-H4 homodimerization and bivalent antigenicity. In determining the affinity kinetics of the antibodies to the free form of B7-H4, both clone #25 and the commercially available clone-H74 showed two-state reaction curves, including rapid dissociation and stable dissociation curves, which can be explained by dimer dissociation and monomer dissociation, respectively (Fig. 3B). Each rapid dissociation rate was equivalent.

B7-H4 ligand instability on the tumor cell surface

B7-H4 expression in the breast cancer cell lines was validated by western blot analysis. Predicted molecular weight of B7-H4 is 29 kDa but it aligns with a size of 50~70 kDa owing to glycosylation and isoform variations (14,26). B7-H4 was detected in cell lysates but not in culture supernatants (Fig. 4A). The soluble-free forms have reportedly been detected in patient sera, but it is difficult to detect in vitro in culture supernatant.

We compared B7-H4 staining in the MDA-MB 468 cell line under suppressive (azide-containing at 4°C) or normal (azide-free at RT) conditions by flow cytometry (Fig. 4B). B7-H4 staining was positive under suppressive conditions using clone #18 and clone #25 antibodies; however, there was no detection under normal conditions. In addition, B7-H4 staining was also not apparent in samples that underwent an overnight incubation at 37°C and 5% CO2.

In vivo experiments

In the MDA-MB-468 transplanted nude mouse model, the strong B7-H4 expression on the tumor cell membrane was observed by immunohistochemistry (IHC), and the intensity of the staining was heterogeneous in the tumor tissue (Fig. 5A). By contrast, the B7-H4-negative NCI-H2170 cell line (control) did not show any B7-H4 expression in tumor tissues. We administered antibody clone #25 (mouse IgG1) to the MDA-MB-468 transplanted mouse model, but no tumor suppressive effect was observed (Fig. 5B).

Indirect ADCC elicited tumor cell death

Based on the observations that i) there was loss of B7-H4 expression on the tumor cell surfaces under usual conditions and ii) there was lack of antitumor activity due to non-functional ADCC in vivo, we employed an indirect ADCC-redirecting T-cell cytotoxicity assay to study B7-H4 using polyclonal anti-mouse IgG-mediated linking of anti-CD3 and anti-B7-H4 antibodies. Freshly enriched T cells indirectly targeting B7-H4 exhibited clone #25-dependent cellular cytotoxicity against the MDA-MB468 cells (Fig. 6). By contrast, clones #18 (mIgG2b), #113 (mIgG1) and #209 (mIgG1) did not induce targeted cell death.

Discussion

A comprehensive whole exome sequencing and gene expression analysis of surgically resected tumor tissues derived from 2,000 patients registered in the HOPE project was performed by the Shizuoka Cancer Center (SCC), Japan over the last 2 years. Focusing on the B7 and CD28 gene families, we found that B7-H4 was altered in several types of cancer (Fig. 1). Unlike other B7 family genes, B7-H4 is also expressed in many normal tissues at the mRNA level. B7-H4 expression in cancer cell lines is low level except for ovarian and breast cancers, while mammary gland epithelium normally expresses B7-H4 (14,20). In vitro-cultured cell lines reflect a similar B7-H4 expression profile as in normal tissues, but expression is not comparable to that of lung cancer tissues in vivo because of B7-H4 modulation.

Our monoclonal antibody clones #113 and #209 bound to B7-H4-transfected HEK293 cells but not to the breast cancer cell line MDA-MB-468 expressing B7-H4 (Figs. 2A and 4B). No B7-H4 gene mutations in the MDA-MB-468 cell line were found, so we hypothesized that the natural expression possibly harbors some type of antibody-binding site as a result of homodimer or heterodimer formation on the cell surface. In addition, we observed that B7-H4 intensity varied as a result of staining temperature and cellular activity (Fig. 4B). Considering that B7-H4 was not identified in culture supernatants (Fig. 4A), tumor cell surface-expressed B7-H4 is likely internalized. The B7-H4 molecule consists of merely two amino acids in the cytoplasm and does not have any signaling function. B7-H4 may be involved in immune regulation as a ligand or decoy ligand for receptors, or it may form a heterodimer with other known B7 family ligands (27). B7-H4 nuclear localization was reported (28), but the expressed isoforms and constructs appear to mainly function on the cell membrane to regulate intercellular signaling.

The studies concerning B7-H4 expression on tumor tissues are not consistent, which may be explained partially by incompatible antibody use (29). However, differences in expression on immune cells suggest immune receptor mobility such as the TCR, CD19 and CD28 families; cross-linking of these antibodies could easily result in receptor internalization on activated lymphocytes (3033). The disappearance of B7-H4 on immune cells may be due to the cross-linking of antibodies (24). Mobility of the B7-H4 ligand on tumor cells was not accounted for (Fig. 4B) and puzzled expression in ovarian cancer (17,18) may explain this phenomenon. This may therefore explain why ADCC could not be performed on tumor cells naturally expressing B7-H4 (Fig. 5B).

The B7-H4 antibody was able to induce cytotoxicity of tumor cells expressing hB7-H4 or mB7-H4 by retroviral vectors through ADCC or complement-dependent cytotoxicity (11), but these observations have not been demonstrated in tumors spontaneously expressing B7-H4. We examined whether the clone #25 Ab could act for MDA-MB-468 tumors transplanted in nude mice, but tumor growth was not suppressed, despite B7-H4 expression in tumors in vivo (Fig. 5A). This is possibly the result of antigen disappearance on the cell surface (Fig. 4B). Cancer therapy targeting immune regulatory molecules are difficult to test in a mouse model, and the benefit of targeting human B7-H4 in human tumors has not yet been shown. In addition, GPI-anchored glycine in mouse B7-H4 is not conserved in humans, and thus B7-H4 may not behave similarly in mice and humans (6). Recently, single-chain Fv fragments (scFv) against hB7-H4 were reported but did not exhibit tumor growth suppression in vivo. Small molecule antibodies such as scFv leak through the kidneys (34-36), and decreased affinity of scFv (37,38) may be incompatible with this inhibitory effect.

Dynamic and unstable cell surface antigens are probably not a target of conventional ADCC activity, but by redirecting T-cell cellular cytotoxicity using specific antibodies recognizing cell surface CD19 antigens instead of a peptide/MHC complex and T-cell clonal specificity (39) have been successful. Clinical studies of chimeric antigen receptor (CAR) T cells transfected with the CD3 gene linked to antigen-specific scFv have been successful and extended further to other tumor targets. Bispecific scFv linked to CD19 and CD3 was also succeeded in clinical study (40).

Here, we used indirect ADCC assays targeting B7-H4 (Fig. 6) and demonstrated a clone #25 Ab-dependent cellular cytotoxicity in breast cancer cells. Since clone #25 (mouse IgG1) does not bind to human Fc receptors, this tumor lysis appears to result from effector T cells but not natural killer cells. Ab clone #18 (IgG2b) and others that bound to B7-H4 on HEK293 cells did not induce breast cancer cell death in this study. These observations are the first to report anti-B7H4 Ab-mediated ADCC activity against tumors spontaneously expressing B7-H4. Therefore, anti-B7H4 Ab-mediated cell death may be considered an alternative therapy for PD-L1-negative and B7H4-positive cancers.

B7-H4 upregulation in lung cancer tissues (Fig. 1) could be induced by cytokines secreted from tumor-infiltrating lymphocytes or by the accumulation of B7-H4-expressing suppressive macrophages derived from abundant intrinsic alveolar-associated macrophages (41,42). In cancer, tumor-infiltrating macrophages and regulatory T cells collectively suppress tumor-associated antigen-specific T cells via B7-H4 and consequently protect tumor cells from immune attacks (18,22). Therefore, approaches to targeting immune suppressor cells expressing B7-H4 are needed for effective cancer therapy, in addition to targeting tumor cells. Elimination of B7-H4-expressing tumor cells and immune-suppressive macrophages by redirecting cytotoxic T cells could be a novel approach for next-generation therapies against cancer.

Abbreviations:

ADCC

antibody-dependent cellular cytotoxicity

B7-H4

B7 family homolog 4

Ab

antibody

Acknowledgments

This study was supported by a grant to Yasuto Akiyama by JSPS KAKENHI (grant no. 25430166), Japan.

References

1 

Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al: Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 363:711–723. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al: Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 369:134–144. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 366:2455–2465. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al: Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 348:124–128. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Sica GL, Choi IH, Zhu G, Tamada K, Wang SD, Tamura H, Chapoval AI, Flies DB, Bajorath J and Chen L: B7-H4, a molecule of the B7 family, negatively regulates T cell immunity. Immunity. 18:849–861. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Prasad DV, Richards S, Mai XM and Dong C: B7S1, a novel B7 family member that negatively regulates T cell activation. Immunity. 18:863–873. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Zang X, Loke P, Kim J, Murphy K, Waitz R and Allison JP: B7x: a widely expressed B7 family member that inhibits T cell activation. Proc Natl Acad Sci USA. 100:10388–10392. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Chen L: Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 4:336–347. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12:252–264. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Choi IH, Zhu G, Sica GL, Strome SE, Cheville JC, Lau JS, Zhu Y, Flies DB, Tamada K and Chen L: Genomic organization and expression analysis of B7-H4, an immune inhibitory molecule of the B7 family. J Immunol. 171:4650–4654. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Jeon H, Vigdorovich V, Garrett-Thomson SC, Janakiram M, Ramagopal UA, Abadi YM, Lee JS, Scandiuzzi L, Ohaegbulam KC, Chinai JM, et al: Structure and cancer immunotherapy of the B7 family member B7x. Cell Reports. 9:1089–1098. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Krambeck AE, Thompson RH, Dong H, Lohse CM, Park ES, Kuntz SM, Leibovich BC, Blute ML, Cheville JC and Kwon ED: B7-H4 expression in renal cell carcinoma and tumor vasculature: Associations with cancer progression and survival. Proc Natl Acad Sci USA. 103:10391–10396. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Miyatake T, Tringler B, Liu W, Liu SH, Papkoff J, Enomoto T, Torkko KC, Dehn DL, Swisher A and Shroyer KR: B7-H4 (DD-O110) is overexpressed in high risk uterine endometrioid adenocarcinomas and inversely correlated with tumor T-cell infiltration. Gynecol Oncol. 106:119–127. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Simon I, Zhuo S, Corral L, Diamandis EP, Sarno MJ, Wolfert RL and Kim NW: B7-h4 is a novel membrane-bound protein and a candidate serum and tissue biomarker for ovarian cancer. Cancer Res. 66:1570–1575. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Thompson RH, Zang X, Lohse CM, Leibovich BC, Slovin SF, Reuter VE, Cheville JC, Blute ML, Russo P, Kwon ED, et al: Serum-soluble B7x is elevated in renal cell carcinoma patients and is associated with advanced stage. Cancer Res. 68:6054–6058. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Rahbar R, Lin A, Ghazarian M, Yau HL, Paramathas S, Lang PA, Schildknecht A, Elford AR, Garcia-Batres C, Martin B, et al: B7-H4 expression by nonhematopoietic cells in the tumor microenvironment promotes antitumor immunity. Cancer Immunol Res. 3:184–195. 2015. View Article : Google Scholar

17 

Zang X, Sullivan PS, Soslow RA, Waitz R, Reuter VE, Wilton A, Thaler HT, Arul M, Slovin SF, Wei J, et al: Tumor associated endothelial expression of B7-H3 predicts survival in ovarian carcinomas. Mod Pathol. 23:1104–1112. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Kryczek I, Zou L, Rodriguez P, Zhu G, Wei S, Mottram P, Brumlik M, Cheng P, Curiel T, Myers L, et al: B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J Exp Med. 203:871–881. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Kryczek I, Wei S, Zhu G, Myers L, Mottram P, Cheng P, Chen L, Coukos G and Zou W: Relationship between B7-H4, regulatory T cells, and patient outcome in human ovarian carcinoma. Cancer Res. 67:8900–8905. 2007. View Article : Google Scholar : PubMed/NCBI

20 

National Cancer Institute: Genotype-Tissue Expression (GTEx). http://www.gtexportal.org/home. 2015

21 

Broad institute: The Cancer Cell Line Encyclopedia (CCLE). https://www.broadinstitute.org/ccle/home. 2015

22 

Dangaj D, Lanitis E, Zhao A, Joshi S, Cheng Y, Sandaltzopoulos R, Ra HJ, Danet-Desnoyers G, Powell DJ Jr and Scholler N: Novel recombinant human b7-h4 antibodies overcome tumoral immune escape to potentiate T-cell antitumor responses. Cancer Res. 73:4820–4829. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Abadi YM, Jeon H, Ohaegbulam KC, Scandiuzzi L, Ghosh K, Hofmeyer KA, Lee JS, Ray A, Gravekamp C and Zang X: Host b7x promotes pulmonary metastasis of breast cancer. J Immunol. 190:3806–3814. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Lee JS, Scandiuzzi L, Ray A, Wei J, Hofmeyer KA, Abadi YM, Loke P, Lin J, Yuan J, Serreze DV, et al: B7x in the periphery abrogates pancreas-specific damage mediated by self-reactive CD8 T cells. J Immunol. 189:4165–4174. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Workman P, Aboagye EO, Balkwill F, Balmain A, Bruder G, Chaplin DJ, Double JA, Everitt J, Farningham DAH, Glennie MJ, et al: Guidelines for the welfare and use of animals in cancer research. Br J Cancer. 102:1555–1577. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Tringler B, Zhuo S, Pilkington G, Torkko KC, Singh M, Lucia MS, Heinz DE, Papkoff J and Shroyer KR: B7-h4 is highly expressed in ductal and lobular breast cancer. Clin Cancer Res. 11:1842–1848. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Butte MJ, Keir ME, Phamduy TB, Sharpe AH and Freeman GJ: Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 27:111–122. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Zhang L, Wu H, Lu D, Li G, Sun C, Song H, Li J, Zhai T, Huang L, Hou C, et al: The costimulatory molecule B7-H4 promote tumor progression and cell proliferation through translocating into nucleus. Oncogene. 32:5347–5358. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Smith JB, Stashwick C and Powell DJ Jr: B7-H4 as a potential target for immunotherapy for gynecologic cancers: A closer look. Gynecol Oncol. 134:181–189. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Pulczynski S, Boesen AM and Jensen OM: Antibody-induced modulation and intracellular transport of CD10 and CD19 antigens in human B-cell lines: An immunofluorescence and immunoelectron microscopy study. Blood. 81:1549–1557. 1993.PubMed/NCBI

31 

Gerber HP, Kung-Sutherland M, Stone I, Morris-Tilden C, Miyamoto J, McCormick R, Alley SC, Okeley N, Hayes B, Hernandez-Ilizaliturri FJ, et al: Potent antitumor activity of the anti-CD19 auristatin antibody drug conjugate hBU12-vcMMAE against rituximab-sensitive and -resistant lymphomas. Blood. 113:4352–4361. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Schaffar L, Dallanegra A, Breittmayer JP, Carrel S and Fehlmann M: Monoclonal antibody internalization and degradation during modulation of the CD3/T-cell receptor complex. Cell Immunol. 116:52–59. 1988. View Article : Google Scholar : PubMed/NCBI

33 

Boyer C, Auphan N, Gabert J, Blanc D, Malissen B and Schmitt-Verhulst AM: Comparison of phosphorylation and internalization of the antigen receptor/CD3 complex, CD8, and class I MHC-encoded proteins on T cells. Role of intracytoplasmic domains analyzed with hybrid CD8/class I molecules. J Immunol. 143:1905–1914. 1989.PubMed/NCBI

34 

He J, Wang Y, Feng J, Zhu X, Lan X, Iyer AK, Zhang N, Seo Y, VanBrocklin HF and Liu B: Targeting prostate cancer cells in vivo using a rapidly internalizing novel human single-chain antibody fragment. J Nucl Med. 51:427–432. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Willuda J, Kubetzko S, Waibel R, Schubiger PA, Zangemeister-Wittke U and Plückthun A: Tumor targeting of mono-, di-, and tetravalent anti-p185HER-2 miniantibodies multimerized by self-associating peptides. J Biol Chem. 276:14385–14392. 2001.PubMed/NCBI

36 

Deyev SM, Waibel R, Lebedenko EN, Schubiger AP and Plückthun A: Design of multivalent complexes using the barnase·barstar module. Nat Biotechnol. 21:1486–1492. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Pleckaityte M, Mistiniene E, Lasickiene R, Zvirblis G and Zvirbliene A: Generation of recombinant single-chain antibodies neutralizing the cytolytic activity of vaginolysin, the main virulence factor of Gardnerella vaginalis. BMC Biotechnol. 11:1002011. View Article : Google Scholar : PubMed/NCBI

38 

Willuda J, Honegger A, Waibel R, Schubiger PA, Stahel R, Zangemeister-Wittke U and Plückthun A: High thermal stability is essential for tumor targeting of antibody fragments: Engineering of a humanized anti-epithelial glycoprotein-2 (epithelial cell adhesion molecule) single-chain Fv fragment. Cancer Res. 59:5758–5767. 1999.PubMed/NCBI

39 

Porter DL, Levine BL, Kalos M, Bagg A and June CH: Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 365:725–733. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, Noppeney R, Viardot A, Hess G, Schuler M, et al: Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 321:974–977. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Richters A, Sherwin RP and Richters V: The lymphocyte and human lung cancers. Cancer Res. 31:214–222. 1971.PubMed/NCBI

42 

Cohen AB and Cline MJ: The human alveolar macrophage: Isolation, cultivation in vitro, and studies of morphologic and functional characteristics. J Clin Invest. 50:1390–1398. 1971. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 36 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Iizuka A, Kondou R, Nonomura C, Ashizawa T, Ohshima K, Kusuhara M, Isaka M, Ohde Y, Yamaguchi K, Akiyama Y, Akiyama Y, et al: Unstable B7-H4 cell surface expression and T-cell redirection as a means of cancer therapy. Oncol Rep 36: 2625-2632, 2016
APA
Iizuka, A., Kondou, R., Nonomura, C., Ashizawa, T., Ohshima, K., Kusuhara, M. ... Akiyama, Y. (2016). Unstable B7-H4 cell surface expression and T-cell redirection as a means of cancer therapy. Oncology Reports, 36, 2625-2632. https://doi.org/10.3892/or.2016.5084
MLA
Iizuka, A., Kondou, R., Nonomura, C., Ashizawa, T., Ohshima, K., Kusuhara, M., Isaka, M., Ohde, Y., Yamaguchi, K., Akiyama, Y."Unstable B7-H4 cell surface expression and T-cell redirection as a means of cancer therapy". Oncology Reports 36.5 (2016): 2625-2632.
Chicago
Iizuka, A., Kondou, R., Nonomura, C., Ashizawa, T., Ohshima, K., Kusuhara, M., Isaka, M., Ohde, Y., Yamaguchi, K., Akiyama, Y."Unstable B7-H4 cell surface expression and T-cell redirection as a means of cancer therapy". Oncology Reports 36, no. 5 (2016): 2625-2632. https://doi.org/10.3892/or.2016.5084