1
|
Fang J, Qian JJ, Yi S, Harding TC, Tu GH, VanRoey M, Jooss K. Stable antibody expression at therapeutic levels using the 2A peptide. Nat Biotechnol 2005; 23:584-90. [PMID: 15834403 DOI: 10.1038/nbt1087] [Citation(s) in RCA: 306] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Accepted: 03/10/2005] [Indexed: 11/09/2022]
Abstract
Therapeutic monoclonal antibodies (mAbs) are currently being developed for the treatment of cancer and other diseases. Despite clinical success, widespread application of mAb therapies may be limited by manufacturing capabilities. In this paper, we describe a mAb delivery system that allows continuous production of a full-length antibody at high-concentrations in vivo after gene transfer. The mAb is expressed from a single open reading frame by linking the heavy and light chains with a 2A self-processing peptide derived from the foot-and-mouth disease virus. Using this expression system, we generated a recombinant adeno-associated virus vector encoding the VEGFR2-neutralizing mAb DC101 (rAAV8-DC101). A single dose of rAAV8-DC101 resulted in long-term expression of >1,000 microg/ml of DC101 in mice, demonstrating significant anti-tumor efficacy. This report describes the first feasible gene therapy approach for stable delivery of mAbs at therapeutic levels, which may serve as an attractive alternative to direct injection of mAbs.
Collapse
|
Journal Article |
20 |
306 |
2
|
Chen S, Lee LF, Fisher TS, Jessen B, Elliott M, Evering W, Logronio K, Tu GH, Tsaparikos K, Li X, Wang H, Ying C, Xiong M, VanArsdale T, Lin JC. Combination of 4-1BB agonist and PD-1 antagonist promotes antitumor effector/memory CD8 T cells in a poorly immunogenic tumor model. Cancer Immunol Res 2014; 3:149-60. [PMID: 25387892 DOI: 10.1158/2326-6066.cir-14-0118] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunotherapies targeting the programmed death 1 (PD-1) coinhibitory receptor have shown great promise for a subset of patients with cancer. However, robust and safe combination therapies are still needed to bring the benefit of cancer immunotherapy to broader patient populations. To search for an optimal strategy of combinatorial immunotherapy, we have compared the antitumor activity of the anti-4-1BB/anti-PD-1 combination with that of the anti-PD-1/anti-LAG-3 combination in the poorly immunogenic B16F10 melanoma model. Pronounced tumor inhibition occurred only in animals receiving anti-PD-1 and anti-4-1BB concomitantly, while combining anti-PD-1 with anti-LAG-3 led to a modest degree of tumor suppression. The activity of the anti-4-1BB/anti-PD-1 combination was dependent on IFNγ and CD8(+) T cells. Both 4-1BB and PD-1 proteins were elevated on the surface of CD8(+) T cells by anti-4-1BB/anti-PD-1 cotreatment. In the tumor microenvironment, an effective antitumor immune response was induced as indicated by the increased CD8(+)/Treg ratio and the enrichment of genes such as Cd3e, Cd8a, Ifng, and Eomes. In the spleen, the combination treatment shaped the immune system to an effector/memory phenotype and increased the overall activity of tumor-specific CD8(+) CTLs, reflecting a long-lasting systemic antitumor response. Furthermore, combination treatment in C57BL/6 mice showed no additional safety signals, and only minimally increased severity of the known toxicity relative to 4-1BB agonist alone. Therefore, in the absence of any cancer vaccine, anti-4-1BB/anti-PD-1 combination therapy is sufficient to elicit a robust antitumor effector/memory T-cell response in an aggressive tumor model and is therefore a candidate for combination trials in patients.
Collapse
|
Journal Article |
11 |
213 |
3
|
Lin J, Lalani AS, Harding TC, Gonzalez M, Wu WW, Luan B, Tu GH, Koprivnikar K, VanRoey MJ, He Y, Alitalo K, Jooss K. Inhibition of lymphogenous metastasis using adeno-associated virus-mediated gene transfer of a soluble VEGFR-3 decoy receptor. Cancer Res 2005; 65:6901-9. [PMID: 16061674 DOI: 10.1158/0008-5472.can-05-0408] [Citation(s) in RCA: 211] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The presence of metastases in regional lymph nodes is a strong indicator of poor patient survival in many types of cancer. It has recently been shown that the lymphangiogenic growth factor, vascular endothelial growth factor-C (VEGF-C), and its receptor, VEGF receptor-3 (VEGFR3), may play a pivotal role in the promotion of metastasis to regional lymph nodes. In this study, human prostate and melanoma tumor models that preferentially metastasize to the lymph nodes following s.c. tumor cell implantation were established from lymph node metastases via in vivo selection. Melanoma tumor cell sublines established from lymph node metastasis express higher amounts of VEGF-C than the parental tumor cells. The inhibition of tumor-derived VEGF-C with a soluble VEGFR3 decoy receptor, sVEGFR3-Fc, expressed via a recombinant adeno-associated viral vector, potently blocks tumor-associated lymphangiogenesis and tumor metastasis to the lymph nodes, when the treatment was initiated before the tumor implantation. In addition, sVEGFR3-Fc serum levels required for efficient blockade of lymph node metastases are strictly dependent on the VEGF-C levels generated by the primary tumor. Recombinant adeno-associated virus-mediated gene transfer of sVEGFR3-Fc may represent a feasible therapeutic strategy for blockade of lymphogenous metastasis.
Collapse
|
Journal Article |
20 |
211 |
4
|
Li B, Lalani AS, Harding TC, Luan B, Koprivnikar K, Huan Tu G, Prell R, VanRoey MJ, Simmons AD, Jooss K. Vascular endothelial growth factor blockade reduces intratumoral regulatory T cells and enhances the efficacy of a GM-CSF-secreting cancer immunotherapy. Clin Cancer Res 2007; 12:6808-16. [PMID: 17121902 DOI: 10.1158/1078-0432.ccr-06-1558] [Citation(s) in RCA: 187] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose of the present study was to evaluate granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy in combination with vascular endothelial growth factor (VEGF) blockage in preclinical models. EXPERIMENTAL DESIGN Survival and immune response were monitored in the B16 melanoma and the CT26 colon carcinoma models. VEGF blockade was achieved by using a recombinant adeno-associated virus vector expressing a soluble VEGF receptor consisting of selected domains of the VEGF receptors 1 and 2 (termed sVEGFR1/R2). Dendritic cell and tumor infiltrating lymphocyte activation status and numbers were evaluated by fluorescence-activated cell sorting analysis. Regulatory T cells were quantified by their CD4+CD25hi and CD4+FoxP3+ phenotype. RESULTS The present study established that GM-CSF-secreting tumor cell immunotherapy with VEGF blockade significantly prolonged the survival of tumor-bearing mice. Enhanced anti-tumor protection correlated with an increased number of activated CD4+ and CD8+ tumor-infiltrating T cells and a pronounced decrease in the number of suppressive regulatory T cells residing in the tumor. Conversely, overexpression of VEGF from tumors resulted in elevated numbers of regulatory T cells in the tumor, suggesting a novel mechanism of VEGF-mediated immune suppression at the tumor site. CONCLUSION GM-CSF-secreting cancer immunotherapy and VEGF blockade increases the i.t. ratio of effector to regulatory T cells to provide enhanced antitumor responses. This therapeutic combination may prove to be an effective strategy for the treatment of patients with cancer.
Collapse
MESH Headings
- Animals
- Apoptosis
- Carcinoma/therapy
- Cell Count
- Colonic Neoplasms/therapy
- Combined Modality Therapy
- Dendritic Cells/cytology
- Fas Ligand Protein/physiology
- Gene Expression Regulation, Neoplastic
- Genetic Therapy/methods
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use
- Immunotherapy/methods
- Lymphocytes, Tumor-Infiltrating/cytology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/mortality
- Neoplasms, Experimental/therapy
- Receptors, Vascular Endothelial Growth Factor/genetics
- Receptors, Vascular Endothelial Growth Factor/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Recombinant Proteins/therapeutic use
- Survival Analysis
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/drug effects
- Treatment Outcome
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/metabolism
- fas Receptor/analysis
Collapse
|
Journal Article |
18 |
187 |
5
|
Fang J, Yi S, Simmons A, Tu GH, Nguyen M, Harding TC, VanRoey M, Jooss K. An antibody delivery system for regulated expression of therapeutic levels of monoclonal antibodies in vivo. Mol Ther 2007; 15:1153-9. [PMID: 17375065 DOI: 10.1038/sj.mt.6300142] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Monoclonal antibody (mAb) delivery by gene transfer in vivo may be an attractive alternative to current mAb therapies for applications that require long-term therapy. This article describes a transfer system that allows inducible high-level expression of unmodified mAbs in vivo. A recombinant adeno-associated viral (rAAV) vector is used that comprises an expression cassette consisting of a dimerizer-regulated promoter that drives expression of the antibody heavy and light chains linked by a 2A self-processing peptide and a furin cleavage site. Following intravenous injection of the rAAV vector, serum mAb levels >1 mg/ml were attained by administration of the inducer, rapamycin. Antibody expression could be rapidly shut off by discontinuing treatment with rapamycin. By optimizing the furin cleavage sequence, this system generated native antibody in vivo, decreasing the likelihood of a host immune response to foreign sequences. In summary, this optimized mAb expression system allows regulated high-level expression of native full-length mAbs in vivo and may offer a new opportunity for delivery of therapeutic mAbs in the clinic.
Collapse
|
Journal Article |
18 |
83 |
6
|
Lee LF, Axtell R, Tu GH, Logronio K, Dilley J, Yu J, Rickert M, Han B, Evering W, Walker MG, Shi J, de Jong BA, Killestein J, Polman CH, Steinman L, Lin JC. IL-7 promotes T(H)1 development and serum IL-7 predicts clinical response to interferon-β in multiple sclerosis. Sci Transl Med 2012; 3:93ra68. [PMID: 21795588 DOI: 10.1126/scitranslmed.3002400] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The interleukin-7 receptor α chain (IL-7Rα) gene was identified as a top non-major histocompatibility complex-linked risk locus for multiple sclerosis (MS). Recently, we showed that a T helper 1 (T(H)1)-driven, but not a T(H)17-driven, form of MS exhibited a good clinical response to interferon-β (IFN-β) therapy. We now demonstrate that high serum levels of IL-7, particularly when paired with low levels of IL-17F, predict responsiveness to IFN-β and hence a T(H)1-driven subtype of MS. We also show that although IL-7 signaling is neither necessary nor sufficient for the induction or expansion of T(H)17 cells, IL-7 can greatly enhance both human and mouse T(H)1 cell differentiation. IL-7 alone is sufficient to induce human T(H)1 differentiation in the absence of IL-12 or other cytokines. Furthermore, targeting IL-7/IL-7Rα is beneficial in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Mice treated with IL-7Rα-blocking antibodies before or after onset of paralysis exhibited reduced clinical signs of EAE, with reduction in peripheral naïve and activated T cells, whereas central memory T, regulatory T, B, and natural killer cell populations were largely spared. IL-7Rα antibody treatment markedly reduced lymphocyte infiltration into the central nervous system in mice with EAE. Thus, a serum profile of high IL-7 may signify a T(H)1-driven form of MS and may predict outcome in MS patients undergoing IFN-β therapy. Blockade of IL-7 and the IL-7Rα pathway may have therapeutic potential in MS and other autoimmune diseases.
Collapse
|
Research Support, Non-U.S. Gov't |
13 |
80 |
7
|
Harding TC, Koprivnikar KE, Tu GH, Zayek N, Lew S, Subramanian A, Sivakumaran A, Frey D, Ho K, VanRoey MJ, Nichols TC, Bellinger DA, Yendluri S, Waugh J, McArthur J, Veres G, Donahue BA. Intravenous administration of an AAV-2 vector for the expression of factor IX in mice and a dog model of hemophilia B. Gene Ther 2004; 11:204-13. [PMID: 14712305 DOI: 10.1038/sj.gt.3302142] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previous experiments have demonstrated the stable expression of factor IX (FIX) protein in mice and canine models of hemophilia B following portal vein gene transfer with a recombinant adeno-associated virus (rAAV) vector encoding FIX. Here, we present the results of studies that further optimized the rAAV vector transgene cassette used to express FIX and explored the use of the less-invasive intravenous (i.v.) route of vector administration for the treatment of hemophilia B. First, a liver-specific promoter was evaluated in conjunction with cis-acting regulatory elements in mice. Constructs that included both the beta-globin intron and the woodchuck hepatitis virus post-transcriptional regulatory element resulted in the highest level of FIX expression in vivo. Using this optimized vector, we demonstrate that i.v. injection was feasible for hepatic gene transfer in mice, achieving 70-80% of portal vein expression levels of FIX. In further studies using the Chapel Hill strain of hemophilia B dogs, we demonstrate for the first time FIX expression and partial correction of the bleeding disorder following i.v. administration of an AAV vector.
Collapse
|
Journal Article |
21 |
43 |
8
|
Strasser AW, Selk R, Dohmen RJ, Niermann T, Bielefeld M, Seeboth P, Tu GH, Hollenberg CP. Analysis of the alpha-amylase gene of Schwanniomyces occidentalis and the secretion of its gene product in transformants of different yeast genera. EUROPEAN JOURNAL OF BIOCHEMISTRY 1989; 184:699-706. [PMID: 2806251 DOI: 10.1111/j.1432-1033.1989.tb15069.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We have cloned and characterized the alpha-amylase gene (AMY1) of the yeast Schwanniomyces occidentalis. A cosmid gene library of S. occidentalis DNA was screened in Saccharomyces cerevisiae for alpha-amylase secretion. The positive clone contained a DNA fragment harbouring an open reading frame of 1536 nucleotides coding for a 512-amino-acid polypeptide with a calculated Mr of 56,500. The deduced amino acid sequence reveals significant similarity to the sequence of the Saccharomycopsis fibuligera and Aspergillus oryzae alpha-amylases. The AMY l gene was found to be expressed from its original promoter in S. cerevisiae, Kluyveromyces lactis and Schizo-saccharomyces pombe leading to an active secreted gene product and thus enabling the different yeast transformants to grow on starch as a sole carbon source.
Collapse
|
|
36 |
38 |
9
|
Nguyen MC, Tu GH, Koprivnikar KE, Gonzalez-Edick M, Jooss KU, Harding TC. Antibody responses to galectin-8, TARP and TRAP1 in prostate cancer patients treated with a GM-CSF-secreting cellular immunotherapy. Cancer Immunol Immunother 2010; 59:1313-23. [PMID: 20499060 PMCID: PMC11030960 DOI: 10.1007/s00262-010-0858-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 04/20/2010] [Indexed: 12/23/2022]
Abstract
A critical factor in clinical development of cancer immunotherapies is the identification of tumor-associated antigens that may be related to immunotherapy potency. In this study, protein microarrays containing >8,000 human proteins were screened with serum from prostate cancer patients (N = 13) before and after treatment with a granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting whole cell immunotherapy. Thirty-three proteins were identified that displayed significantly elevated (P
Collapse
|
research-article |
15 |
20 |
10
|
Zhang P, Tu GH, Wei J, Santiago P, Larrabee LR, Liao-Chan S, Mistry T, Chu MLH, Sai T, Lindquist K, Long H, Chaparro-Riggers J, Salek-Ardakani S, Yeung YA. Ligand-Blocking and Membrane-Proximal Domain Targeting Anti-OX40 Antibodies Mediate Potent T Cell-Stimulatory and Anti-Tumor Activity. Cell Rep 2020; 27:3117-3123.e5. [PMID: 31189099 DOI: 10.1016/j.celrep.2019.05.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/16/2019] [Accepted: 05/06/2019] [Indexed: 12/21/2022] Open
Abstract
Agonistic antibodies targeting the tumor necrosis factor (TNF) superfamily of co-stimulatory receptors (TNFRSF) are progressing through various stages of clinical development for cancer treatment, but the desired and defining features of these agents for optimal biological activity remain controversial. One idea, based on recent studies with CD40, is that non-ligand-blocking antibodies targeting membrane-distal cysteine-rich domain 1 (CRD1) have superior agonistic activities compared with ligand-blocking antibodies targeting more membrane-proximal CRDs. Here, we determined the binding and functional characteristics of a panel of antibodies targeting CRDs 1-4 of OX40 (also known as TNFRSF4 or CD134). In striking contrast to CD40, we found that ligand-blocking CRD2-binding and membrane-proximal CRD4-binding anti-OX40 antibodies have the strongest agonistic and anti-tumor activities. These findings have important translational implications and further highlight that the relationship between epitope specificity and agonistic activity will be an important issue to resolve on a case-by-case basis when optimizing antibodies targeting different co-stimulatory tumor necrosis factor receptors (TNFRs).
Collapse
|
Research Support, Non-U.S. Gov't |
5 |
15 |
11
|
Farina S, Yang H, Tu GH, Gamelin EC, Lin JC, Wang C, Feldman R, Panowski S, Oderup C. Abstract LB-194: Targeting tumor associated myeloid cells with CCR2 inhibitor PF-04136309 enhances gemcitabine/paclitaxel and doxorubicin anti-tumor activity. Cancer Res 2017. [DOI: 10.1158/1538-7445.am2017-lb-194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: MDSCs and macrophages have been reported to be increased in the tumor microenvironment by chemotherapy treatment. These cells may limit the beneficial effects of chemotherapy by limiting drug access or by suppressing the tumor targeted immune response evoked by the chemotherapy. The purpose of the current study was to investigate the ability of chemokine receptor CCR2 inhibitor, PF-04136309, to block tumor MDSC and macrophage accumulation and to investigate the combination of this treatment with standard of care chemotherapies. Experimental design: The ability of CCR2 inhibitor, PF-04136309, to reduce tumor growth in mice was evaluated using orthotopically or SC injected syngeneic colon, pancreatic and ovarian tumor cell lines. PF-04136309 was evaluated as single agent, in combination with doxorubicin, or in the combination of gemcitabine and paclitaxel. Results: In a model of ovarian carcinomatosis (ID8), where tumor progression is driven by MDSCs and macrophages, single agent PF-04136309 reduced peritoneal tumor load as well as the total volume and rate of ascites fluid production. Even delayed treatment of established tumors, at 6 weeks post tumor inoculation, was effective. Ascites volume correlated with overall tumor load in treated mice. The treatment of ovarian tumor bearing mice with PF-04136309 in combination with doxorubicin, resulted in significant reduction in tumor load and ascites formation, beyond that produced by doxorubicin alone. To evaluate the addition of PF-04136309 to the chemotherapy combination, gemcitabine/paclitaxel, we selected models of pancreatic (Pan02) and colon (MC38) carcinoma, where PF-04136309 did not show single agent efficacy. The addition of PF-04136309 resulted in significantly decreased tumor burden compared to mice treated with chemotherapy alone. To investigate the mechanism of action of PF-04136309 we selected the MC38 model. PF-04136309 treatment of MC38 tumor bearing mice resulted in a substantial reduction, as a proportion of CD45+ cells, of MDSCs (CD11b+MHCII-Ly6C+) in the circulation (80%), and of MDSCs (60%) and macrophages (CD11b+F4/80+MHCII+Ly6C/G-) (70%) in the tumor, respectively. Conclusion: PF-04136309 significantly improved the response to several chemotherapeutics, providing strong rationale for CCR2 antagonism as an addition to standard of care chemotherapies. PF-04136309 is currently being evaluated in combination with gemcitabine/nab-paclitaxel in metastatic pancreatic cancer patients. The observed inhibitory effect of PF-04136309 on ID8 ascites formation, the synergy with doxorubicin, and the known intervention of MDSC and macrophages in human ovarian peritoneal carcinomatosis, suggests potential clinical efficacy with reduction of ascites volume and synergy in combination with pegylated doxorubicin in platinum resistant ovarian cancer patients.
Citation Format: Sasha Farina, Hsunhui Yang, Guang Huan Tu, Erick C. Gamelin, John C. Lin, Changyu Wang, Reid Feldman, Siler Panowski, Cecilia Oderup. Targeting tumor associated myeloid cells with CCR2 inhibitor PF-04136309 enhances gemcitabine/paclitaxel and doxorubicin anti-tumor activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-194. doi:10.1158/1538-7445.AM2017-LB-194
Collapse
|
|
8 |
|