The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Nov. 12, 2015, is named 19540_0104 PTWO_SL.txt and is 11,000 bytes in size.
This invention relates to the fields of cancer treatment and vaccine mediated therapies.
Of the seven most common cancers in the US, melanoma is the only one to have increasing incidence in the 20 century, with an estimated 76,100 incident cases of invasive melanoma diagnosed in the US in 2014, and 9,710 estimated deaths. Additionally, melanoma is very aggressive. According to the American Cancer Society, once the cancer progresses to stage IIIB, the ten-year survival rate dips below 50%, with that rate plummeting to 10-15% for stage IV. Although melanoma is highly immunogenic, the anti-inflammatory tumor microenvironment greatly inhibits any immune-based intervention. The discovery of immune checkpoint inhibitors targeting tumor microenvironment interactions with effector T-cells has marked a major discovery and paradigm shift in research and treatment modalities. The success of ipilimumab (anti-cytotoxic T-lymphocyte-associated antigen 4 [αCTLA-4]) in the clinic has paved the way for competing anti-Programmed Cell Death 1 (αPD-1) drugs pembrolizumab and nivolumab. These antibodies directly target proteins on the surface of effector T cells linked to signaling pathways that inhibit T cell activation. Melanoma generates a tolerogenic environment that is active at stages that precede generation of effector T cells.
Another promising line of investigation is that of cancer vaccines. A novel DNA vaccine platform has been developed that includes the chemokine macrophage inflammatory protein 3α (MIP3α/CCL20) fused to the melanoma-associated antigen GP100. This platform is superior to standard DNA vaccines because the chemokine targets nascent protein to the immature dendritic cells (iDCs) that are pertinent to the development of an adaptive immune response. The iDCs process antigen via both class I and class II pathways, jump-starting both humoral and cell-mediated immunity. Studies in a malaria challenge system have demonstrated that combining this iDC targeting vaccine construct with an adjuvant results in resistance to infection that is improved by orders of magnitude, compared to either adjuvant or vaccine construct alone. The aspects of the malaria challenge system were presented in U.S. Pat. No. 8,557,248, which is herein incorporated by reference. This vaccine platform has also been shown to work in malaria with circumsporozoite protein antigen, in melanoma prophylactically with gp100 antigen, and in lymphoma therapeutically with OFA-iLRP antigen. Preliminary data show that this construct is efficacious in a therapeutic setting with melanoma, prolonging median survival by 29%.
Previous melanoma therapies have provided hope for better treatments but other treatments have failed because later stage melanomas eventually escape the effect of the treatment. This failure can be attributable to mutations arising in the proteins targeted by the immune response or by mechanisms of immune tolerance that downregulate the immune response to tumor antigens, which are frequently proteins normally expressed, albeit at lower frequency, on non-malignant cells. While some current therapies seek to counteract some of the tolerance mechanisms, only a minority of patients respond to such interventions.
There is a need for the development of, and improvement of vaccines for the treatment of melanoma.
The invention provides compositions and methods for treatment of various cancers. Vaccine constructs are provided comprising a cytokine fused to a cancer antigen. The present invention provides DNA and protein vaccine constructs, which can be based on the fusion of a cytokine, e.g., MIP-3α, and a melanoma-associated antigen, e.g., GP100. The vaccine constructs of the present invention can be provided for use in combination with various adjuvants, and various other cancer therapies. The present invention provides for combining the vaccine constructs with anti-IL-10, for example.
The present invention also provides methods of making and using the DNA and protein vaccine constructs in the treatment of melanoma. Also provided are methods for using the protein vaccine constructs in Antibody-Coupled T-cell Receptor (“ACTR”) technology.
In certain embodiments, multiple facets of the tumor system are simultaneously targeted. These treatments may have separate modes of action, and their combination may provide a synergistic action to dramatically improve patient treatment. αCTLA-4 may be combined with granulocyte-macrophage colony-stimulating factor (GM-CSF), which can increase overall survival while decreasing toxicity. In another embodiment, a recombinant adenovirus vaccine was given in combination with αPD-1 and α4-1BB (CD137), which elicited melanoma remission in mice. In certain embodiments, combination therapies including αCTLA-4 and αPD-1, have improved outcomes.
In a murine embodiment, a dendritic cell vaccine with GP100 antigen was combined with αIL-10. Certain embodiments may not utilize problematic virus vectors or expensive and technically demanding adoptive dendritic cell transfers. Further, the present invention is to provide a level of specific immunity to protect against development of recurrences or metastases, which would not be the case for combinations of antibody and chemotherapies.
The addition of therapeutic antibodies αIL-10 and αPD-1 to certain embodiments is to produce a synergistic interaction between a vaccine construct and countermeasures to address tumor-initiated immunosuppression and to modulate or completely suppress tumor growth and spread; thus, prolonging median survival by 47%. Combination treatments may include agents that affect multiple facets of tumorigenesis, shrinking primary tumors while building vaccination-induced immunity to eliminate metastases and prevent relapses.
In certain embodiments, tumor microenvironment immune parameters can act as non-invasive corollaries of protection in a combination therapy system. In certain embodiments, a melanoma model with IL-10 knockout mice on the C57Bl/6 background is utilized. In certain embodiments, the knockout mouse in combination with therapeutic vaccination of MIP-3α-GP100 to analyze local immune parameters can be utilized. The present invention provides, in part, a focus on levels of pro- and anti-inflammatory cytokines such as Interferon-γ, Tumor Necrosis Factor-α, Transforming Growth Factor-β, etc.; counts of different immune cell types, especially Tregs and CD8+ T-cells; and percentage of GP100-specific Tumor Infiltrating Lymphocytes (TILs).
The effect of immunological interventions on the tumor microenvironment can be characterized to identify corollaries of effective therapy. Using methods similar to those described in U.S. Pat. No. 8,557,248 B2, herein incorporated by reference, qRT-PCR can be used to identify alterations in the cytokine/chemokine environment within the tumor, as well as examine by flow cytometry the composition of immune cell populations within the tumor. Focus may be placed on an array of cytokines including, but not limited to IFN-γ, TNF-α, TGF-β, IL-2, IL-6, IL-10, and IL-17. Flow cytometry can be employed to quantify the presence of different immune cell types, especially regulatory T cells (Treg) and CD8+ T-cells, and percentage of GP100-specific Tumor Infiltrating Lymphocytes (TILs).
Certain embodiments can utilize melanoma combination therapy utilizing therapeutic antibodies and MIP-3α-antigen fusion vaccination. Some embodiments can utilize melanoma-associated antigens in the same plasmid or in separate plasmids fused to MIP-3α. Complimentary antibodies in addition to αIL-10 and αPD-1 can be added to the therapy to enhance protective effects. The synergistic efficacy of blocking other checkpoint inhibitors and activating effector T-cells can be improved using the embodiments.
After in vitro analysis, the model can be tested in a prophylactic setting. The standard mouse model of melanoma in young female C57Bl/6 mice can be utilized.
Certain embodiments can demonstrate efficacy in a clinical therapeutic model, where treatments do not begin until after tumor is induced.
A mouse model is useful for studying the impact of immunomodulation on tumor progression and survival. In an effort to balance having sufficient tumor mass for analysis without extensive necrosis, the immune environment in tumors at different time points can be characterized. The optimal time point for tissue examination may differ for groups receiving different or no therapeutic interventions, but coverage of a range of time points should provide important insights into the kinetics and magnitude of the responses associated with different intervention strategies. The initial groups to be compared can be those described in
Having defined the local immune parameters associated with initial interventions, those interventions can be supplemented by using an additional antigen and additional antibodies (summarized in Table 1) to determine their impact on tumor growth, survival, and on the parameters outlined above. The impact of an additional antigen to the vaccine regimen may be incorporated into various embodiments. Utilizing standard bacterial cloning procedures, DNA encoding the clinically relevant antigen tryosinase-related protein 2 (TRP-2) can be inserted into a separate plasmid fused to MIP-3α, as was done for gp100 (
Following alteration of a vaccine construct, different therapeutic antibodies can be tested for synergistic efficacy within this system, beginning with a αIL-10 and αPD1 regimen. For the clinical setting, it may be impractical to administer αIL-10 at the tumor site, especially in the context of metastatic disease. The use of systemic αIL-10 monoclonal antibody may demonstrate no significant toxicity following daily administration over a 21-day interval at a dose of 0.25 mg/kg. To evaluate the potential efficacy of systemic depletion of IL-10, tumor size and survival among three αIL-10 treatment groups may be compared: mice treated with the vaccine construct plus intravenous administration of αIL-10 at doses that range between those used in the human studies to doses that have been used previously in mice (5-250 μg/mouse), mice receiving the previously used local injections at the tumor site, and IL-10 knockout mice. Subsequent experiments will add α4-1BB (CD137), an antibody which acts as an agonist for CD8+ T cell activation and may synergistically inhibit tumor growth when combined with vaccine and αPD1. An embodiment incorporating this regimen can avoid the adverse events that are commonly associated in high frequency with αCTLA4, which can be used in combination with αPD1. The addition of αCTLA4 to the regimen can also be used in the clinical setting. Appropriate controls with and without the vaccine construct and different combinations of the antibodies can be included in all of the comparison studies.
The described regimens may produce complete remission, thus allowing further study to be performed on the mice two months after initial challenge. These mice can be challenged again in the opposite flank and/or intravenously to assess protection from relapse or metastases. These alterations to the protocol can lead to a therapy to greatly alter the course of clinical disease. All data can be statistically analyzed by ANOVA and log-rank tests, as described in preliminary results and as discussed above.
In one embodiment utilizing a therapy combining MIP-3α-antigen fusion DNA vaccines with immunomodulatory antibodies can have potent effects against melanoma in the mouse model and in human patients. Mechanistic immunological correlations can be utilized to fully assess an optimized therapy. An embodiment that induces established tumors to undergo remission by establishment of immunity to multiple antigens via vaccination and by reversal of the anti-immunity tumor microenvironment via a cocktail of immunomodulatory therapeutic antibodies can be utilized. This has great potential clinical impact, because the treatment could not only increase patient short-term outcomes, but could also help prevent metastases and long-term relapses.
The following is a table of adjuvants organized by class and with examples for the melanoma vaccine described herein. These adjuvants will aid in obtaining a high antibody concentration, including use of adjuvants with a protein formulation.
One embodiment of the present invention is the use of a melanoma vaccine construct described for the DNA vaccine but expressing the DNA in bacteria as a protein. It should be understood that the protein can also be expressed in yeast, insect cells or mammalian cells. One strength of this approach is this vaccine is readily adaptable to the appearance of new cancer antigens, termed neoantigens, which arise as a result of ongoing mutations of tumor genes. Therefore, the present invention includes a DNA vaccine framework into which a tumor antigen is inserted and the expression product can be recognized by the immune system. The approach of identifying neoantigens is described in Castle et al. (2012), Exploiting the Mutanome for Tumor Vaccination, Cancer Research; 72(5); 1081-91. While Castle et al. emphasizes neoantigen identification by CD8+ T cells, the present invention is adapted to recognize and identify new tumor proteins that are recognized by antibody and also CD4+ T cells with a separate screening.
As provided in more detail below, the present invention provides for standalone DNA vaccine constructs, standalone protein vaccine constructs, or even combinations of the two. In certain embodiments, the DNA vaccine can be the MIP-3α-melanoma-associated antigen fusion construct used in combination with anti-interleukin-10 (“anti-IL-10”). In other embodiments, the protein formulation of a vaccine construct can be the MIP-3α-melanoma-associated antigen fusion construct in combination with an adjuvant as described herein. The protein formulation of the vaccine construct can be the MIP-3α-melanoma-associated antigen fusion construct in combination with an adjuvant and/or anti-IL-10.
By way of example, several compositions and therapies were used to compare post-induction tumor size and survival of individuals post-tumor induction. This study utilized exclusively 6-8 week old female C57BL/6 mice ordered from Charles River Laboratories (Wilmington, Mass.). Mice were challenged in the left flank subcutaneously with a lethal dose (5×104 cells) of B16F10 melanoma. Tumor size was recorded as square mm, representing length×width (opposing axes) measured by calipers every 1-3 days. The mice were kept in the study until one of the following occured: mouse death, tumor size eclipsing 20 mm in any direction, or extensive tumor necrosis and ulceration. Anti-IL-10 antibody 150 ug/injection; BioXcell JES5.2A51 was administered subcutaneously at the challenge/tumor site beginning day 5 post tumor challenge and continuing once every 3 days for a total of 6 doses. The vaccination plasmid extracted from E. coli using Qiagen® EndoFree® Plasmid Maxi and Giga Kits were used. DNA verified by gel electrophoresis, restriction enzyme analysis, Nanodrop® spectrophotometry, and full insert sequencing. The vaccine comprised solely of purified plasmid DNA encoding MIP-3α-gp100 fusion sequence in endotoxin-free PBS. Mock vaccinations were comprised of endotoxin-free PBS only. DNA injections were administered into the hind leg tibialis muscle. Immediately following injection, the muscle was pulsed using an ECM 830 Electro Square Porator (BTX Harvard Apparatus®) with the following parameters: 106V; 20 ms pulse length; 200 ms pulse interval; 8 total pulses. Vaccinations of 50 ug/dose delivered at days 3, 10, and 17 post tumor challenge. For survival studies, groups included 22-29 mice encompassing 3-4 independent experiments. For analysis of tumor size, groups included 22-47 mice across 4-9 independent experiments, and analysis of day 17 specifically included 22-35 mice per group across 4-6 independent experiments. Tumor size analyses were statistically tested by one-way anova with bonferonni correction. Mouse survival studies were statistically tested by the log-rank test. α<0.05.
For the comparison, melanoma tumors were introduced to animals in the laboratory. The animals were divided into groups for control group (mock), anti-IL-10 treatment group, DNA MIP-3α-GP100 vaccine construct treatment group, and DNA MIP-3α-GP100 vaccine construct plus anti-IL-10 treatment group. As seen in
In
TG*TTGCCTCTCGTACATACAGACGCCTCTTCCTTCCAGAGCTATTGTG
GGTTTCACAAGACAGATGGCCGATGAAGCTTGTGACATTAATGCTATCA
TCTTTCACACGAAGAAAAGAAAATCTGTGTGCGCTGATCCAAAGCAGAA
CTGGGTGAAAAGGGCTGTGAACCTCCTCAGCCTAAGAGTCAAGAAGATG
GAATTCAACGACGCTCAGGCGCCGAAGAGTCTCGAGGCTAGA
AAAGTAC
CCAGAAACCAGGACTGGCTTGGTGTCTCAAGGCAACTCAGAACCAAAGC
CTGGAACAGGCAGCTGTATCCAGAGTGGACAGAAGCCCAGAGACTTGAC
TGCTGGAGAGGTGGTCAAGTGTCCCTCAAGGTCAGTAATGATGGGCCTA
CACTGATTGGTGCAAATGCCTCCTTCTCTATTGCCTTGAACTTCCCTGG
AAGCCAAAAGGTATTGCCAGATGGGCAGGTTATCTGGGTCAACAATACC
ATCATCAATGGGAGCCAGGTGTGGGGAGGACAGCCAGTGTATCCCCAGG
AAACTGACGATGCCTGCATCTTCCCTGATGGTGGACCTTGCCCATCTGG
CTCTTGGTCTCAGAAGAGAAGCTTTGTTTATGTCTGGAAGACCTGGGGC
CAATACTGGCAAGTTCTAGGGGGCCCAGTGTCTGGGCTGAGCATTGGGA
CAGGCAGGGCAATGCTGGGCACACACACCATGGAAGTGACTGTCTACCA
TCGCCGGGGATCCCGGAGCTATGTGCCTCTTGCTCATTCCAGCTCAGCC
TTCACCATTACTGACCAGGTGCCTTTCTCCGTGAGCGTGTCCCAGTTGC
GGGCCTTGGATGGAGGGAACAAGCACTTCCTGAGAAATTCTAGAGAGAT
Internal annotations are described herein below.
In
TPLPSRAIVGFTRQMADEACDINAIIFHTKKRKSVCADPKQNWVKRAVN
LLSLRVKKM
EFNDAQAPKSLEAR
KVPRNQDWLGVSRQLRTKAWNRQLYP
EWTEAQRLDCWRGGQVSLKVSNDGPTLIGANASFSIALNFPGSQKVLPD
GQVIWVNNTIINGSQVWGGQPVYPQETDDACIFPDGGPCPSGSWSQKRS
FVYVWKTWGQYWQVLGGPVSGLSIGTGRAMLGTHTMEVTVYHRRGSRSY
VPLAHSSSAFTITDQVPFSVSVSQLRALDGGNKHFLRNLERSAE{EQKL
Internal annotations are described herein below.
In
G*TTGCCTCTCGTACATACAGACGCCTCTTCCTTCCAGAGCTATTGTGG
GTTTCACAAGACAGATGGCCGATGAAGCTTGTGACATTAATGCTATCAT
CTTTCACACGAAGAAAAGAAAATCTGTGTGCGCTGATCCAAAGCAGAAC
TGGGTGAAAAGGGCTGTGAACCTCCTCAGCCTAAGAGTCAAGAAGATG
G
AATTCAACGACGCTCAGGCGCCGAAGAGTCTCGAGGCTAGA
AAAGTACC
CAGAAACCAGGACTGGCTTGGTGTCTCAAGGCAACTCAGAACCAAAGCC
TGGAACAGGCAGCTGTATCCAGAGTGGACAGAAGCCCAGAGACTTGACT
GCTGGAGAGGTGGTCAAGTGTCCCTCAAGGTCAGTAATGATGGGCCTAC
ACTGATTGGTGCAAATGCCTCCTTCTCTATTGCCTTGAACTTCCCTGGA
AGCCAAAAGGTATTGCCAGATGGGCAGGTTATCTGGGTCAACAATACCA
TCATCAATGGGAGCCAGGTGTGGGGAGGACAGCCAGTGTATCCCCAGGA
AACTGACGATGCCTGCATCTTCCCTGATGGTGGACCTTGCCCATCTGGC
TCTTGGTCTCAGAAGAGAAGCTTTGTTTATGTCTGGAAGACCTGGGGCC
AATACTGGCAAGTTCTAGGGGGCCCAGTGTCTGGGCTGAGCATTGGGAC
AGGCAGGGCAATGCTGGGCACACACACCATGGAAGTGACTGTCTACCAT
CGCCGGGGATCCCGGAGCTATGTGCCTCTTGCTCATTCCAGCTCAGCCT
TCACCATTACTGACCAGGTGCCTTTCTCCGTGAGCGTGTCCCAGTTGCG
GGCCTTGGATGGAGGGAACAAGCACTTCCTGAGAAATCTAGAGAGATCC
Internal annotations are described herein below.
In
PSRAIVGFTRQMADEACDINAIIFHTKKRKSVCADPKQNWVKRAVNLLS
LRVKKM
EFNDAQAPKSLEAR
KVPRNQDWLGVSRQLRTKAWNRQLYPEWT
EAQRLDCWRGGQVSLKVSNDGPTLIGANASFSIALNFPGSQKVLPDGQV
IWVNNTIINGSQVWGGQPVYPQETDDACIFPDGGPCPSGSWSQKRSFVY
AHSSSAFTITDQVPFSVSVSQLRALDGGNKHFLRNLERSAE{EQKLISE
Internal annotations are described herein below.
In
TTCATCCTAAATTTATTGTGGGCTTCACACGGCAGCTGGCCAATGAAGG
CTGTGACATCAATGCTATCATCTTTCACACAAAGAAAAAGTTGTCTGTG
TGCGCAAATCCAAAACAGACTTGGGTGAAATATATTGTGCGTCTCCTCA
GTAAAAAAGTCAAGAACATG-3′.
Internal annotations are described herein below.
In
KLSVCANPKQTWVKYIVRLLSKKVKNM-COO2.
Internal annotations are described herein below.
As shown in SEQ ID NOs: 1-6 above, there are various internal annotations, which are described in the following key:
Key:
As discussed herein, the present invention provides for standalone DNA vaccine constructs, standalone protein vaccine constructs, and combinations thereof. As provided herein, the sequences of certain DNA and protein vaccine construct embodiments are provided. While certain methods of expressing the desired vaccine constructs are disclosed, other methods for expressing the desired vaccine constructs are known. Delivery methods for the DNA and protein vaccine constructs are known. These include plasmid DNA delivery methods of: parenteral delivery (e.g., injection, gene gun, pneumatic (jet) injection); topical application; and cytofectin-mediated delivery.
Administration of the protein vaccine construct of the present invention to an individual in need of melanoma therapeutic care is expected to yield high concentrations of antibodies in the subject. The high concentrations of antibodies created by the protein vaccine construct of the current invention can be used in combination with T cells to develop a chimeric antigen receptor type (CAR T) system. A recently described technology employed for cancer immunotherapy uses T cells carrying antibodies on their surface to target antigens on tumor cells (Prapa, et al. (2015), A novel anti-GD2/4-1BB chimeric antigen receptor triggers neuroblastoma cell killing. Oncotarget 6: 24884-24894; and Kudo, et al. (2014), T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer research 74: 93-103). Essentially, T cells obtained from patients are engineered to express receptors for antibody on their surface. Termed “Antibody-Coupled T-cell Receptor (ACTR) technology”, this technology relies on an engineered T-cell component that can bind antibodies and use them to target the T-cells. When modified T-cells are put back into the patient, they can be targeted to attack tumors by co-administering cancer-specific antibodies. Patents covering the ACTR concept have been filed by St. Jude Children's Research Hospital and the National University of Singapore (U.S. Pat. No. 8,399,645).
The success of this technology is dependent on establishing high concentrations of antibody specific for a tumor antigen. This is applicable to the vaccine constructs of the present invention due to the high levels of antibodies resulting from administration of the vaccine constructs of this invention. While monoclonal antibodies have been used initially in studies of this technology, the ability to rapidly elicit high concentrations of antibodies to antigens for which monoclonal antibodies are not available and particularly for neoantigens that appear as tumor cells mutate would greatly enhance the potential efficacy of this approach. The ability of the MIP-3α vaccine platform to be rapidly modified to express antigens of interest and to elicit remarkably high concentrations of specific antibody should enhance the breadth of activity and ultimately the efficacy of this ACTR technology. An individual could be immunized with a vaccine platform/construct expressing the antigens of interest to be followed by infusion of the ACTR engineered T lymphocytes.
A number of embodiments of the invention have been described. Nevertheless, it will understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US15/60371 | 11/12/2015 | WO | 00 |
Number | Date | Country | |
---|---|---|---|
62078225 | Nov 2014 | US |