SELECTING NEOANTIGENS FOR PERSONALIZED CANCER VACCINE

Information

  • Patent Application
  • 20230197192
  • Publication Number
    20230197192
  • Date Filed
    November 05, 2021
    2 years ago
  • Date Published
    June 22, 2023
    a year ago
Abstract
Disclosed herein are methods for selecting one or more tumor-specific neoantigens from a tumor of a subject for a personalized immunogenic composition. Also disclosed herein are methods for treating cancer in a subject in need thereof by administering an immunogenic composition comprising tumor-specific neoantigens selected using the methods disclosed herein.
Description
REFERENCE TO A SEQUENCE LISTING

This application contains a Sequence Listing in computer readable form. The computer readable form is incorporated herein by reference. Said ASCII copy, created on Oct. 28, 2021, is named 146401_091524_SL.txt and is 75,598 bytes in size.


1. BACKGROUND

Cancer is a leading cause of death worldwide accounting for 1 in 4 of all deaths. Siegel et al., CA: A Cancer Journal for Clinicians, 68:7-30 (2018). There were 18.1 million new cancer cases and 9.6 million cancer-related deaths in 2018. Bray et al., CA: A Cancer Journal for Clinicians, 68(6):394-424. There are a number of existing standard of care cancer therapies, including ablation techniques (e.g., surgical procedures and radiation) and chemical techniques (e.g., chemotherapeutic agents). Unfortunately, such therapies are frequently associated with serious risk, toxic side effects, and extremely high costs, as well as uncertain efficacy.


Cancer immunotherapy (e.g., cancer vaccine) has emerged as a promising cancer treatment modality. The goal of cancer immunotherapy is to harness the immune system for selective destruction of cancer while leaving normal tissues unharmed. Traditional cancer vaccines typically target tumor-associated antigens. Tumor-associated antigens are typically present in normal tissues, but overexpressed in cancer. However, because these antigens are often present in normal tissues immune tolerance can prevent immune activation. Several clinical trials targeting tumor-associated antigens have failed to demonstrate a durable beneficial effect compared to standard of care treatment. Li et al., Ann Oncol., 28 (Suppl 12): xii11-xii17 (2017).


Neoantigens represent an attractive target for cancer immunotherapies. Neoantigens are non-autologous proteins with individual specificity. Neoantigens are derived from random somatic mutations in the tumor cell genome and are not expressed on the surface of normal cells. Id. Because neoantigens are expressed exclusively on tumor cells, and thus do not induce central immune tolerance, cancer vaccines targeting cancer neoantigens have potential advantages, including decreased central immune tolerance and improved safety profile. Id.


The mutational landscape of cancer is complex and tumor mutations are generally unique to each individual subject. Most somatic mutations detected by sequencing do not result in effective neoantigens. Only a small percentage of mutations in the tumor DNA, or a tumor cell, are transcribed, translated, and processed into a tumor-specific neoantigen with sufficient accuracy to design a vaccine that is likely to be effective. Further, not all neoantigens are immunogenic. In fact, the proportion of T cells spontaneously recognizing endogenous neoantigens is about 1% to 2%. See, Karpanen et al., Front Immunol., 8:1718 (2017). Moreover, the cost and time associated with the manufacture of neoantigen vaccines is significant.


Thus, it remains a challenge to efficiently and accurately predict, prioritize, and select neoantigen candidates for immunogenic compositions. Accordingly, there is a significant unmet need for an integrated method to characterize tumor genomic material to identify neoantigens, identify which neoantigens are targeted by the immune system, and select which neoantigens are likely to be suitable for effective immunogenic compositions.


2. SUMMARY

This disclosure relates to a novel method for selecting one or more tumor-specific neoantigens from a tumor of a subject for a subject-specific immunogenic composition. The disclosure also relates to methods of treating cancer in a subject in need thereof by administering an immunogenic composition comprising tumor-specific neoantigens selected using the novel approach for selecting tumor-specific neoantigens and formulating an immunogenic composition comprising the selected tumor-specific neoantigens. The approach beings with obtaining sequence data from the tumor. The sequence data is used to obtain data representing a polypeptide sequence of one or more tumor-specific neoantigens. The sequence data may be nucleotide sequence data, polypeptide sequence data, exome sequence data, transcriptome sequence data, or whole genome nucleotide sequence data. The sequence data may be whole exome sequence data, RNA sequence data, whole genome sequence data or combinations thereof. The sequence data may be a combination of whole exome sequence data, RNA sequence data, and whole genome sequence data.


The polypeptide sequence(s) and MHC molecule(s) of the subject are then inputted into a machine-learning platform. The machine-learning platform is used to identify whether tumor-specific neoantigens are immunogenic (e.g., that the one or more tumor-specific neoantigen will elicit an immune response in the subject). Based on these predictions, the machine-learning platform generates a numerical probability score that one or more tumor-specific neoantigens will elicit an immune response in the subject.


The MCH molecule(s) of the subject may be MHC class I molecule and/or an MHC class II molecule. The polypeptide sequence encoding one or more tumor-specific neoantigens may be from short polypeptides. Short polypeptides are typically presented on MCH class I molecules. Alternatively, the polypeptide sequence encoding one or more tumor-specific neoantigens can be from long polypeptides.


The immune response in the subject can include presentation of one or more tumor-specific neoantigens to the tumor cell surface, presentation of one or more tumor-specific neoantigens by one or more MHC molecules on the tumor cell, or that one or more tumor-specific neoantigens is capable of presentation to T cells by antigen presenting cells.


The immune response in the subject can be a CD4+ mediated response or a CD8+ mediated response. Typically, the immune response is either a CD4+ mediated response or a CD8+ mediated response.


A tumor-specific neoantigen with a higher numerical probability score relative to a lower numerical probability score indicates that the tumor-specific neoantigen will elicit a greater immune response in the subject.


RNA expression, preferably mRNA expression, of the one or more tumor-specific neoantigens in a tumor is also quantified to further identify one or more tumor-specific neoantigens that are sufficiently expressed to elicit an immune response in the subject. Then, tumor clones can be optionally characterized to ensure that the tumor-specific neoantigens represent sufficient fraction (e.g., genetic diversity) across the tumor. In embodiments, a suitable tumor-specific neoantigen may represent about 1% of the tumor. In other instances, a suitable tumor-specific neoantigen may represent about 5% of the tumor.


These parameters are used to calculate a tumor-specific neoantigen score for the one or more tumor-specific neoantigen scores. The tumor-specific neoantigen score is used to select tumor-specific neoantigens suitable for formulation of a subject-specific immunogenic composition. A higher tumor-specific neoantigen score relative to a lower tumor-specific neoantigen score indicates that the neoantigen has stronger immunogenicity, and thus more likely to induce a strong immune response and elicit stable therapeutic effects (i.e., more likely to be suitable for an immunogenic composition). In embodiments, at least about 10 tumor-specific neoantigens are selected to formulate the subject-specific immunogenic composition. In embodiments, at least about 20 tumor-specific neoantigens are selected to formulate the subject-specific immunogenic composition.


The methods disclosed herein can further comprise measuring the ability of the one or more tumor-specific neoantigens to induce an autoimmune response to normal tissue. A tumor-specific neoantigen that induces an autoimmune response in normal tissue will have a lower tumor-specific neoantigen score relative to a tumor-specific neoantigen that does not induce an autoimmune response. A tumor-specific neoantigen that induces an autoimmune response will not be selected for the immunogenic composition.


The formulated immunogenic composition may include at least about 10 tumor-specific neoantigens or at least about 20 tumor-specific neoantigens. The tumor-specific neoantigens can be encoded by short polypeptides or by long polypeptides. The immunogenic composition may comprise a nucleotide sequence, a polypeptide sequence, RNA, DNA, a cell, a plasmid, a vector, a dendritic cell, or a synthetic long peptide. The immunogenic composition can further comprise an adjuvant.


This disclosure also relates to methods of treating cancer in a subject in need thereof comprising administering a personalized immunogenic composition comprising one or more tumor specific neoantigens selected using the methods described herein. The methods disclosed herein can be suited for treating any number of cancers. The tumor can be from melanoma, breast cancer, ovarian cancer, prostate cancer, kidney cancer, gastric cancer, colon cancer, testicular cancer, head and neck cancer, pancreatic cancer, brain cancer, B-cell lymphoma, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, T-cell lymphocytic leukemia, bladder cancer, or lung cancer. Preferably, the cancer is melanoma, breast cancer, lung cancer, and bladder cancer.





3. BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a schematic depicting the approach for selecting one or more tumor-specific neoantigens.



FIG. 2 is schematic flow diagram depicting the bioinformatics analysis of next generation sequencing data (input and output).



FIG. 3 is a flow diagram of the module for clonality deconvolutioion





4. DETAILED DESCRIPTION

This disclosure relates to a novel approach for selecting tumor-specific neoantigens with high-accuracy for potent personalized cancer immunogenic compositions (e.g., subject-specific immunogenic compositions). The disclosure also relates to methods of treating cancer in a subject in need thereof by administering an immunogenic composition comprising tumor-specific neoantigens selected using the novel approach for selecting tumor-specific neoantigens and formulating an immunogenic composition comprising the selected tumor-specific neoantigens. The inventors have developed an approach that: 1) sequences the DNA and/or RNA encoding for the polypeptide sequence of one or more neoantigens; 2) determines whether the tumor-specific neoantigen is immunogenic (e.g., whether a neoantigen can elicit an immune response in the subject); 3) determines whether the tumor expresses an amount of neoantigen sufficient to elicit an immune response; and 4) optionally determines whether the neoantigen represents a sufficient fraction of the tumor. Currently available methods rely on MHC binding affinity predictions to rank and select for neoantigens or the probability a neoantigen will be presented by an MHC molecule. These methods do not predict immunogenicity. Moreover, current methods do not have the capability to evaluate all of these factors with high-accuracy.


The approach begins with sequencing the polypeptide sequence of tumor-specific neoantigens obtained from a tumor biopsy. A prediction machine-learning platform is then used to identify which neoantigens are recognized by MHC molecules of the subject. The platform can determine whether the tumor-specific neoantigens are immunogenic (e.g., that the tumor-specific neoantigen will elicit an immune response in the subject). Based on these predictions, the machine-learning platform generates a numerical probability score that the tumor-specific neoantigens will elicit an immune response. RNA expression, preferably, mRNA expression, of tumor-specific neoantigens is also quantified to focus on the tumor-specific neoantigens that are abundantly expressed, such that they will likely elicit an immune response. Then tumor clones are optionally characterized to ensure that the tumor-specific neoantigens represent sufficient genetic diversity across the tumor. These parameters are used to create a tumor-specific neoantigen score for the tumor-specific neoantigens. The tumor-specific neoantigen score is used to select tumor-specific neoantigens suitable for formulation of a personalized vaccine. A higher tumor-specific neoantigen score relative to a lower tumor-specific neoantigen score indicates that the neoantigen has stronger immunogenicity, and thus more likely to induce a strong immune response and elicit stable therapeutic effects.


All publications and patents cited in this disclosure are incorporated by reference in their entirety. To the extent, the material incorporated by reference contradicts or is inconsistent with this specification, the specification will supersede any such material. The citation of any references herein is not an admission that such references are prior art to the present disclosure. When a range of values is expressed, it includes embodiments using any particular value within the range. Further, reference to values stated in ranges includes each and every value within that range. All ranges are inclusive of their endpoints and combinable. When values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment. Reference to a particular numerical value includes at least that particular value, unless the context clearly dictates otherwise. The use of “or” will mean “and/or” unless the specific context of its use dictates otherwise.


Various terms relating to aspects of the description are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein. The techniques and procedures described or referenced herein are generally well understood and commonly employed using conventional methodologies by those skilled in the art, such as, for example, the widely utilized molecular cloning methodologies described in Sambrook et al., Molecular Cloning: A Laboratory Manual 4th ed. (2012) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. As appropriate, procedures involving the use of commercially available kits and reagents are generally carried out in accordance with manufacturer-defined protocols and conditions unless otherwise noted.


As used herein, the singular forms “a,” “an,” and “the” include plural forms unless the context clearly indicates otherwise. The terms “include,” “such as,” and the like are intended to convey inclusion without limitation, unless otherwise specifically indicated.


Unless otherwise indicated, the terms “at least,” “less than,” and “about,” or similar terms preceding a series of elements or a range are to be understood to refer to every element in the series or range. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.


The term “cancer” refers to the physiological condition in subjects in which a population of cells is characterized by uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate and/or certain morphological features. Often cancers can be in the form of a tumor or mass, but may exist alone within the subject, or may circulate in the blood stream as independent cells, such a leukemic or lymphoma cells. The term cancer includes all types of cancers and metastases, including hematological malignancy, solid tumors, sarcomas, carcinomas and other solid and non-solid tumors. Examples of cancers include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers include squamous cell cancer, small cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer (e.g., triple negative breast cancer, Hormone receptor positive breast cancer), osteosarcoma, melanoma, colon cancer, colorectal cancer, endometrial (e.g., serous) or uterine cancer, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulvar cancer, thyroid cancer, hepatic carcinoma, and various types of head and neck cancers. Triple negative breast cancer refers to breast cancer that is negative for expression of the genes for estrogen receptor (ER), progesterone receptor (PR), and Her2/neu. Hormone receptor positive breast cancer refers to breast cancer that is positive for at least one of the following: ER or PR, and negative for Her2/neu (HER2).


The term “neoantigen” as used herein refers to an antigen that has at least one alteration that makes it distinct from the corresponding parent antigen, e.g., via mutation in a tumor cell or post-translational modification specific to a tumor cell. A mutation can include a frameshift, indel, missense or nonsense substitution, splice site alteration, genomic rearrangement or gene fusion, or any genomic expression alteration giving rise to a neoantigen. A mutation can include a splice mutation. Post-translational modifications specific to a tumor cell can include aberrant phosphorylation. Post-translational modifications specific to a tumor cell can also include a proteasome-generated spliced antigen. See, Lipe et al., Science, 354(6310):354:358 (2016). In general, point mutations account for about 95% mutations in tumors and indels and frame-shift mutations account for the rest. See, Snyder et al., N Engl J Med., 371:2189-2199 (2014).


As used herein the term “tumor-specific neoantigen” is a neoantigen present in a subject's tumor cell or tissue, but not in the subject's normal cell or tissue.


The term “next generation sequencing” or “NGS” as used herein refers to sequencing technologies having increased throughput as compared to traditional approaches (e.g., Sanger sequencing), with the ability to generate hundreds of thousands of sequence reads at a time.


The term “neural network” as used herein refers to a machine-learning model for classification or regression consisting of multiple layers of linear transformations followed by element-wise nonlinearities typically trained via stochastic gradient descent and back-propagation.


The term “subject” as used herein refers to any animal, such as any mammal, including but not limited to, humans, non-human primates, rodents, and the like. In some embodiments, the mammal is a mouse. In some embodiments, the mammal is a human.


The term “tumor cell” as used herein refers to any cell that is a cancer cell or is derived from a cancer cell. The term “tumor cell” can also refer to a cell that exhibits cancer-like properties, e.g., uncontrollable reproduction, resistance to anti-growth signals, ability to metastasize, and loss of ability to undergo programed cell death.


Additional description of the methods and guidance for the practice of the methods are provided herein.


I. Methods for Selecting Tumor-Specific Neoantigens


Disclosed herein are methods for selecting tumor-specific neoantigens from a tumor of a subject that are suitable for subject-specific immunogenic compositions. Suitable tumor-specific neoantigens are tumor-specific neoantigens that are likely presented on the cell surface of the tumor, are likely to be immunogenic, are predicted to be expressed in sufficient amounts to elicit an immune response in the subject, and optionally represent sufficient diversity across the tumor.


The first step in selecting one or more tumor-specific neoantigens from a tumor of a subject comprises obtaining sequence data from the tumor. The sequence data is used to obtain data representing a polypeptide sequence of one or more tumor-specific neoantigens. Generally, sequence data representing a polypeptide sequence of one or more tumor-specific neoantigens is determined by subjecting a tumor sample to sequence analysis.


The sequence data can be exome sequence data, transcriptome sequence data, whole genome nucleotide sequence data, nucleotide sequence data, or polypeptide sequence data. The sequence data may be whole exome sequence data, RNA sequence data, whole genome sequence data or combinations thereof. The sequence data may be a combination of whole exome sequence data, RNA sequence data, and whole genome sequence data.


Various methods of obtaining sequence data may be used in the methods described herein. Sequencing methods are well known in the art and include, but are not limited to, PCR-based methods, including real-time PC, whole exome sequencing, deep sequencing, high-throughput sequencing, or combinations thereof. In some embodiments, the foregoing techniques and procedures are performed according to the methods described in e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual 4th ed. (2012) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. See also, Austell et al., Current Protocols in Molecular Biology, ed., Greene Publishing and Wiley-Interscience New York (1992) (with periodic updates).


Sequencing methods may also include, but are not limited to, high-throughput sequencing, single-cell RNA sequence, RNA sequencing, pyrosequencing, sequencing-by synthesis, single-molecule sequencing, nanopore sequencing, semiconductor sequencing, sequencing-by-synthesis, sequencing-by-ligation, sequencing-by-hybridization, RNA-Sew (Illumina), Digital Gene Expression (Helicos), next generation sequencing, Single Molecule Sequencing by Synthesis (SMSS) (Helicos), massively-parallel sequencing, Clonal Single Molecule Array (Solexa), shotgun sequencing, Maxam-Hilbery or Sanger sequencing, whole genome sequencing, whole exome sequencing, primer walking, sequencing using PacBio, SOLid, Ion Torrent, or Nanopore platforms and any other sequencing methods known in the art. The sequencing method employed herein to obtain sequence data is preferably high-throughput sequencing. High-throughput sequencing technologies are capable of sequencing multiple nucleic acid molecules in parallel, enabling millions of nucleic acid molecules to be sequenced at a time. See, Churko et al., Circ. Res. 112(12):1613-1623 (2013).


In some instances, whole exome sequences, RNA sequencing, whole genome sequencing or combinations thereof can be performed. In some instances, a combination of whole exome sequences, RNA sequencing, whole genome sequencing can be performed.


In some cases, high-throughput sequencing can be next generation sequencing. There are a number of different next generation platforms using different sequencing technologies (e.g., using the HiSeq or MiSeq instruments available from Illumina (San Diego, Calif.)). Any of these platforms can be employed for sequencing the genetic material disclosed herein. Next generation sequencing is based on sequencing a large number of independent reads, each representing anywhere between 10 to 1000 bases of nucleic acid. Sequencing by synthesis is a common technique used in next generation sequencing. In general, sequencing involves hybridizing a primer to a template to form a template/primer duplex, contacting the duplex with a polymerase in the presence of a detectably-labeled nucleotide under conditions that permit the polymerase to add nucleotides to the primer in a template-dependent manner. Signal from the detectable label is then used to identify the incorporated base and the steps are sequentially repeated in order to determine the linear order of nucleotides in the template. Exemplary detectable labels include radiolabels, florescent labels, enzymatic labels, etc. Numerous techniques are known for detecting sequences, such as the Illumina NextSeq platform by cycle end sequencing.


Once sequence data representing the polypeptide sequence of one or more tumor specific neoantigens is obtained, the sequence data, along with the MHC molecule of the subject, is inputted into a machine-learning platform. The machine-learning platform generates a numerical probability score that forecasts whether the one or more tumor-specific neoantigens are immunogenic (e.g. will elicit an immune response in the subject).


MHC molecules transport and present peptides on the cell surface. The MHC molecules are classified as MHC molecules of class I and of class II. MHC class I are present on the surface of almost all cells of the body, including most tumor cells. The proteins of MHC class I are loaded with antigens that usually originate from endogenous proteins or from pathogens present inside cells, and are then presented to cytotoxic T-lymphocytes (i.e., CD8+). The MHC class I molecules can comprise HLA-A, HLA-B, or HLA-C. The MHC molecules of class II are only present on dendritic cells, B lymphocytes, macrophages and other antigen-presenting cells. They present mainly peptides, which are processed from external antigen sources, i.e. outside of the cells, to T-helper (Th) cells (i.e., CD4+). The MHC class II molecules can comprise HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1, HLA-DRA, and HLA-DRB1. In some occasions, MHC class II molecules can also be expressed on cancer cells.


MHC class I molecules and/or MHC class II molecules can be inputted into the machine-learning platform. Typically, either MHC class I molecules or MHC class II molecules are inputted into the machine-learning platform. In some embodiments, MHC class I molecules are inputted into the machine-learning platform. In other embodiments, MHC class II molecules are inputted into the machine-learning platform.


MHC class I molecules bind to short peptides. MHC class I molecules can accommodate peptides generally about 8 amino acids to about 10 amino acids in length. In embodiments, the sequence data encoding one or more tumor-specific neoantigens are short peptides about 8 amino acids to about 10 amino acids in length. MHC class II molecules bind to peptides that are longer in length. MHC class II can accommodate peptides which are generally about 13 amino acids in length to about 25 amino acids in length. In embodiments, the sequence data encoding one or more tumor-specific neoantigens are long peptides about 13 to 25 amino acids in length.


The sequence data encoding one or more tumor-specific neoantigens can be about 5 amino acids in length, about 6 amino acids in length, about 7 amino acids in length, about 8 amino acids in length, about 9 amino acids in length, about 10 amino acids in length, about 11 amino acids in length, about 12 amino acids in length, about 13 amino acids in length, about 14 amino acids in length, about 15 amino acids in length, about 16 amino acids in length, about 17 amino acids in length, about 18 amino acids in length, about 19 amino acids in length, about 20 amino acids in length, about 21 amino acids in length, about 22 amino acids in length, about 23 amino acids in length, about 24 amino acids in length, about 25 amino acids in length, about 26 amino acids in length, about 27 amino acids in length, about 28 amino acids in length, about 29 amino acids in length, or about 30 amino acids in length.


The machine-learning platform predicts the likelihood that one or more tumor-specific neoantigens are immunogenic (e.g., will elicit an immune response).


Immunogenic tumor-specific neoantigens are not expressed in normal tissues. They can be presented by antigen-presenting cells to CD4+ and CD8+ T-cells to generate an immune response. In embodiments, an immune response in the subject elicited by the one or more tumor-specific neoantigens comprises presentation of the one or more tumor-specific neoantigens to the tumor cell surface. More specifically, the immune response in the subject elicited by the one or more tumor-specific neoantigens comprises presentation of the one or more tumor-specific neoantigens by one or more MHC molecules on the tumor cell. It is expected that the immune response elicited by the one or more tumor-specific neoantigens is a T-cell mediated response. The immune response in the subject elicited by the one or more tumor-specific neoantigens may involve one or more tumor-specific neoantigens being capable of presentation to T-cells by antigen presenting cells, such as dendritic cells. Preferably, the one or more tumor-specific neoantigens is capable of activating CD8+ T-cells and/or CD4+ T-cells.


In embodiments, the machine-learning platform can predict the likelihood the one or more tumor-specific neoantigens will activate CD8+ T cells. In embodiments, the machine learning platform can predict the likelihood that the one or more tumor-specific neoantigens will activate CD4+ T cells. In some instances, the machine-learning platform can predict the antibody titer that the one or more tumor-specific neoantigens can elicit. In other instances, the machine-learning platform can predict the frequency of CD8+ activation by the one or more tumor-specific neoantigens.


The machine-learning platform can include a model trained on training data. Training data can be obtained from a series of distinct subjects. The training data can comprise data derived from healthy subjects, as well as subjects having cancer. The training data may include various data that can be used to generate a probability score that indicates whether the one or more tumor-specific neoantigens will elicit an immune response in a subject. Exemplary training data can include data representing nucleotide or polypeptide sequences derived from normal tissue and/or cells, data representing nucleotide or polypeptide sequences derived from tumor tissue, data representing MHC peptidome sequences from normal and tumor tissue, peptide-MHC binding affinity measurement, or combinations thereof The reference data can further comprise mass spectrometry data, DNA sequencing data, RNA sequencing data, clinical data from healthy subjects and subjects having cancer, cytokine profiling data, T cell cytotoxicity assay data, peptide-MHC mono-or-multimer data, and proteomics data for single-allele cell lines engineered to express a predetermined MHC allele that are subsequently exposed to synthetic protein, normal and tumor human cell lines, fresh and frozen primary samples, and T-cell assays.


The machine-learning platform can be a supervised learning platform, an unsupervised learning platform, or a semi-supervised learning platform. The machine-learning platform can use sequence-based approach to generate a numerical probability that the one or more tumor-specific neoantigens can elicit an immune response (e.g., will induce a high or low antibody response or CD8+response). Sequence based predictions can include supervised machine-learning modules including, artificial neural networks (e.g., deep or otherwise), support vector machines, K-nearest neighbor, Logistic Multiple Network-constrained Regression (LogMiNeR), regression tree, random forest, adaboost, XGBoost, or hidden Markov models. These platforms require training data sets that include known MHC binding peptides.


Numerous prediction programs have been employed to predict whether a tumor-specific neoantigen can be presented on an MHC molecule and elicit an immune response. Exemplary predictive programs include, for example, HLAminer (Warren et al., Genome Med., 4:95 (2012); HLA type predicted by orienting the assembly of shotgun sequence data and comparing it with the reference allele sequence database), VariantEffect Predictor Tool (McLaren et al., Genome Biol., 17:122 (2016)), NetMHCpan (Andreatta et al., Bioinformatics., 32:511-517 (2016); sequence comparison method based on artificial neural network, and predict the affinity of peptide-MHC-I type molecular), UCSC browser (Kent et al., Genome Res., 12:996-1006 (2002)), CloudNeo pipeline (Bais et al., Bioinformatics, 33:3110-2 (2017)), OptiType (Szolek et al., Bioinformatics, 30:3310-316 (2014)), ATHLATES (Liu C et al., Nucleic Acids Res. 41:e142 (2013)), pVAC-Seq (Handal et al., Genome Med. 8:11 (2016), MuPeXI (Bjerregaard et al., Cancer Immunol Immunother., 66:1123-30 (2017)), Strelka (Saunders et al., Bioinformatics. 28:1811-7 (2012)), Strelka2 (Kim et al., Nat Methods. 2018;15:591-4.), VarScan2 (Koboldt et al., Genome Res., 22:568-76 (2012)), Somaticseq (Fang L et al., Genome Biol., 16:197 (2015)), SMMPMBEC (Kim et al., BMC Bioinformatics., 10:394 (2009)), NeoPredPipe (Schenck RO, BMC Bioinformatics., 20:264 (2019)), Weka (Witten et al., Data mining: practical machine-learning tools and techniques. 4th ed. Elsevier, ISBN: 97801280435578 (eBook) (2017), or Orange (Demsar et al., Orange: Data Mining Toolbox in Python., J. Mach Learn Res., 14:2349-2353 (2013). Any known predictive programs may be employed as the machine-learning platform to generate a numerical probability score that indicates whether the neoantigen will elicit an immune response.


Depending on the machine-learning platform employed, additional filters can be applied to prioritize tumor-specific neoantigen candidates, including: elimination of hypothetical (Riken) proteins; use of an antigen processing algorithm to eliminate epitopes that are not likely to be proteolytically produced by the constitutive- or immune-proteasome and prioritization of neoantigens where the neoantigen has a higher predicted binding affinity than the corresponding wildtype sequence.


The numerical probability score can be a number between 0 and 1. In embodiments, the numerical probability score can be a number of 0, 0.0001, 0.0002, 0.0003, 0.0004, 0.0005, 0.0006, 0.0007, 0.0008, 0.0009, 0.001, 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.20, 0.30, 0.40, 0.50, 0.60, 0.70, 0.80, 0.90, or 1. A tumor-specific neoantigen with a higher numerical probability score relative to a lower numerical probability score indicates that the tumor-specific neoantigen will elicit a greater immune response in the subject, and thus is likely to be a suitable candidate for an immunogenic composition. For example, a tumor-specific neoantigen with a numerical probability score of 1 will likely elicit a greater immune response in a subject than a tumor-specific neoantigen having a numerical probability score of 0.05. Similarly, a tumor-specific neoantigen having a numerical probability score of 0.5 will likely elicit a greater immune response in a subject than a tumor-specific neoantigen with a numerical probability score of 0.1.


A higher numerical probability score relative to a lower numerical probability score is preferable. Preferably, tumor-specific neoantigen having a numerical probability score of at least 0.8, 0.81, 0.82. 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.9, 0.95, 0.96, 0.97, 0.98, 0.99, or 1 indicates that an immune response will likely be elicited in the subject.


While a higher numerical probability score is preferable, a lower numerical probability score may still indicate that the tumor-specific neoantigen is capable of eliciting a sufficient immune response, such that the tumor-specific neoantigen is likely to be a suitable candidate.


In instances, the machine-learning platform described herein can also predict the likelihood that the one or more tumor-specific neoantigens will be presented by a MHC molecule on a tumor cell. The machine-learning platform can predict the likelihood that one or more tumor-specific neoantigens will be presented by a MHC class I molecule or MHC class II molecule.


The methods for selecting one or more tumor-specific neoantigens may further comprise a step of measuring, in silico, the affinity of one or more tumor-specific neoantigens to bind to a MHC molecule in the subject. A tumor-specific neoantigen that has a binding affinity with a MHC molecule of less than about 1000 nM indicates that the one or more tumor-specific neoantigens may be suitable for an immunogenic composition. A tumor-specific neoantigen that has a binding affinity with a MHC molecule of less than about 500 nM, of less than about 400 nM, of less than about 300 nM, of less than about 200 nM, of less than about 100 nM, of less than about 50 nM can indicate that one or more tumor-specific neoantigens may be suitable for an immunogenic composition. The affinity of the one or more tumor-specific neoantigens to bind to a MHC molecule in the subject can predict tumor-specific neoantigen immunogenicity. Alternatively, median affinity can be an effective way to predict tumor-specific neoantigen immunogenicity. Median affinity can be calculated using epitope prediction algorithms, such as NetMHCpan, ANN, SMM and SMMPMBEC.


RNA expression of one or more tumor-specific neoantigens is also quantified. RNA expression of one or more tumor-specific neoantigens is quantified to identify one or more neoantigens that will elicit an immune response in a subject. A variety of methods exist for measuring RNA expression. Known techniques, which may measure RNA expression, include RNA-seq, and in situ hybridization (e.g., FISH), Northern blot, DNA microarray, Tiling array, and quantitative polymerase chain reaction (qPCR). Other known techniques in the art can be used to quantify RNA expression. RNA can be messenger RNA (mRNA), short-interfering RNA (siRNA), microRNA (miRNA), circular RNA (circRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), small nucleolar RNA (snRNA), Piwi-interacting RNA (piRNA), long non-coding RNA (long ncRNA), sub-genomic RNA (sgRNA), RNA from integrating or non-integrating viruses, or any other RNA. Preferably, mRNA expression is measured.


The methods disclosed herein can optionally comprise sequencing tumor clones. Tumor clones are sequenced to identify one or more tumor-specific neoantigens that represent a sufficient fraction of the tumor. Tumor clones can be sequenced, for example, using the sequence techniques disclosed herein and using other known sequencing technologies known by those skilled in the art.


In embodiments, a tumor-specific neoantigen that has a tumor clone fraction of at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, or at least about 30% across the tumor indicates that the tumor-specific neoantigen represents a sufficient fraction of the tumor. A sufficient fraction of the tumor indicates that the tumor-specific neoantigen provides sufficient genetic diversity across the tumor.


The method can further comprise measuring the ability of the one or more tumor-specific neoantigen to induce an autoimmune response in normal tissues. It is expected that a tumor-specific neoantigen that has similar sequence to a normal antigen may induce an autoimmune response in normal tissue. For example, a tumor-specific neoantigen that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% similar to a normal antigen may induce an autoimmune response. Tumor-specific neoantigens that are predicted to induce an autoimmune response are not prioritized for the immunogenic composition. Tumor-specific neoantigens that are predicted to induce an autoimmune response are typically not selected for the immunogenic composition. The method can further comprise measuring the ability of the one or more tumor-specific neoantigen to invoke immunological tolerance. Tumor-specific neoantigens that are predicted to invoke immunological tolerance are not prioritized for the immunogenic composition. Tumor-specific neoantigens that are predicted to invoke immunological tolerance are not prioritized for the immunogenic composition.


A tumor-specific score is calculated based on the data generated by obtaining a numerical probability score that the one or more tumor-specific neoantigens will elicit an immune response in the subject and the RNA expression levels of the one or more tumor-specific neoantigens. The tumor clone fraction across the tumor can optionally be included in addition to the above calculation used to calculate the tumor-specific score. A tumor-specific neoantigen that has a high numerical probability score (e.g., the tumor-specific neoantigen is immunogenic) and has a high level of RNA expression, will be prioritized. In comparison, a tumor-specific antigen that is predicted to induce an autoimmune response will have a lower tumor-specific neoantigen score relative to a tumor-specific neoantigen that does not induce an immune response and will not be selected for inclusion in an immunogenic composition. A tumor-specific neoantigen that has a high numerical probability score (e.g., the tumor-specific neoantigen is immunogenic), has a high level of RNA expression, and provides a sufficient tumor clone fraction across the tumor will be prioritized.


A tumor-specific neoantigen that has a high numerical probability score (e.g., the tumor-specific neoantigen is immunogenic) and optionally provides a sufficient tumor clone fraction across the tumor, but has low levels of RNA expression, will have a lower tumor-specific score in comparison to a tumor-specific neoantigen that has a high numerical probability score, high RNA expression levels, and optionally provides sufficient tumor clone fraction across the tumor. In this example, the tumor-specific neoantigen with a lower tumor-specific score will not be prioritized over the tumor-specific neoantigen with the higher tumor-specific score. A tumor-specific neoantigen that has a high numerical probability score (e.g., the tumor-specific neoantigen is immunogenic) and has sufficient levels of RNA expression to elicit an immune response, but does not provide a sufficient tumor clone fraction across the tumor will have a lower tumor-specific score in comparison to a tumor-specific neoantigen that has a high numerical probability score, high RNA expression levels, and provides sufficient tumor clone fraction across the tumor. A tumor-specific neoantigen that has sufficient levels of RNA expression to elicit an immune response, provides a sufficient tumor fraction across the tumor, but has a low numerical probability score, will have a lower tumor-specific score in comparison to a tumor-specific neoantigen that has a high numerical probability score, high RNA expression levels, and provides a sufficient tumor fraction across the tumor.


Finally, one or more tumor-specific neoantigens based on the tumor-specific score are selected for formulation of a subject-specific immunogenic composition. In embodiments, at least about 1, at least about 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, at least about 10, at least about 11, at least about 12, at least about 13, at least about 14, at least about 15, at least about 16, at least about 17, at least about 18, at least about 19, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 50 or more tumor-specific neoantigens are selected for the immunogenic composition. Typically, at least about 10 tumor-specific neoantigens are selected. In other instances, at least about 20 tumor-specific neoantigens are selected.


II. Methods of Treating


This disclosure also relates to methods of treating cancer in a subject in need thereof comprising administering a personalized immunogenic composition comprising one or more tumor specific neoantigens selected using the methods described herein.


The cancer can be any solid tumor or any hematological tumor. The methods disclosed herein are preferably suited for solid tumors. The tumor can be a primary tumor (e.g., a tumor that is at the original site where the tumor first arose). Solid tumors can include, but are not limited to, breast cancer tumors, ovarian cancer tumors, prostate cancer tumors, lung cancer tumors, kidney cancer tumors, gastric cancer tumors, testicular cancer tumors, head and neck cancer tumors, pancreatic cancer tumors, brain cancer tumors, and melanoma tumors. Hematological tumors can include, but are not limited to, tumors from lymphomas (e.g., B cell lymphomas) and leukemias (e.g., acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, and T cell lymphocytic leukemia).


The methods disclosed herein can be used for any suitable cancerous tumor, including hematological malignancy, solid tumors, sarcomas, carcinomas, and other solid and non-solid tumors. Illustrative suitable cancers include, for example, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), adrenocortical carcinoma, anal cancer, appendix cancer, astrocytoma, basal cell carcinoma, brain tumor, bile duct cancer, bladder cancer, bone cancer, breast cancer, bronchial tumor, carcinoma of unknown primary origin, cardiac tumor, cervical cancer, chordoma, colon cancer, colorectal cancer, craniopharyngioma, ductal carcinoma, embryonal tumor, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, fibrous histiocytoma, Ewing sarcoma, eye cancer, germ cell tumor, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, gestational trophoblastic disease, glioma, head and neck cancer, hepatocellular cancer, histiocytosis, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumor, Kaposi sarcoma, kidney cancer, Langerhans cell histiocytosis, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ, lung cancer, macroglobulinemia, malignant fibrous histiocytoma, melanoma, Merkel cell carcinoma, mesothelioma, metastatic squamous neck cancer with occult primary, midline tract carcinoma involving NUT gene, mouth cancer, multiple endocrine neoplasia syndrome, multiple myeloma, mycosis fungoides, myelodysplastic syndrome, myelodysplastic/myeloproliferative neoplasm, nasal cavity and par nasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-small cell lung cancer, oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytomas, pituitary tumor, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell cancer, renal pelvis and ureter cancer, retinoblastoma, rhabdoid tumor, salivary gland cancer, Sezary syndrome, skin cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, spinal cord tumor, stomach cancer, T-cell lymphoma, teratoid tumor, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, urethral cancer, uterine cancer, vaginal cancer, vulvar cancer, and Wilms tumor. Preferably, the cancer is melanoma, breast cancer, ovarian cancer, prostate cancer, kidney cancer, gastric cancer, colon cancer, testicular cancer, head and neck cancer, pancreatic cancer, brain cancer, B-cell lymphoma, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, T-cell lymphocytic leukemia, bladder cancer, or lung cancer. Melanoma is of particular interest. Breast cancer, lung cancer, and bladder cancer are also of particular interest.


Immunogenic compositions stimulate a subject's immune system, especially the response of specific CD8+ T cells or CD4+ T cells. Interferon gamma produced by CD8+ and T helper CD4+ cells regulate the expression of PD-L1. PD-L1 expression in tumor cells is upregulated when attacked by T cells. Therefore, tumor vaccines may induce the production of specific T cells and simultaneously upregulate the expression of PD-L1, which may limit the efficacy of the immunogenic composition. In addition, while the immune system is activated, the expression of T cell surface reporter CTLA-4 is correspondingly increased, which binds with the ligand B7-1/B7-2 on antigen-presenting cells and plays an immunosuppressant effect. Thus, in some instances, the subject may further be administered an anti-immunosuppressive or immunostimulatory, such as a checkpoint inhibitor. Checkpoint inhibitors can include, but are not limited to, anti-CTL4-A antibodies, anti-PD-1 antibodies and anti-PD-L1 antibodies. These checkpoint inhibitors bind to the immune checkpoint proteins of T cells to remove the inhibition of T cell function by tumor cells. Blockade of CTLA-4 or PD-L1 by antibodies can enhance the immune response to cancerous cells in the patient. CTLA-4 has been shown effective when following a vaccination protocol.


An immunogenic composition comprising one or more tumor-specific neoantigens can be administered to a subject that has been diagnosed with cancer, is already suffering from cancer, has recurrent cancer (i.e., relapse), or is at risk of developing cancer. An immunogenic composition comprising one or more tumor-specific neoantigens can be administered to a subject that is resistant to other forms of cancer treatment (e.g., chemotherapy, immunotherapy, or radiation). An immunogenic composition comprising one or more tumor-specific neoantigens can be administered to the subject prior to other standard of care cancer therapies (e.g., chemotherapy, immunotherapy, or radiation). An immunogenic composition comprising one or more tumor-specific neoantigens can be administered to the subject concurrently, after, or in combination to other standard of care cancer therapies (e.g., chemotherapy, immunotherapy, or radiation).


The subject can be a human, dog, cat, horse, or any animal for which a tumor specific response is desired.


The immunogenic composition is administered to the subject in an amount sufficient to elicit an immune response to the tumor-specific neoantigen and to destroy, or at least partially arrest, symptoms and/or complications. In embodiments, the immunogenic composition can provide a long-lasting immune response. A long-lasting immune response can be established by administering a boosting dose of the immunogenic composition to the subject. The immune response to the immunogenic composition can be extended by administering to the subject a boosting dose. In embodiments, at least one, at least two, at least three or more boosting doses can be administered to abate the cancer. A first boosting dose may increase the immune response by at least 50%, at least 100%, at least 200%, at least 300%, at least 400%, at least 500%, or at least 1000%. A second boosting dose may increase the immune response by at least 50%, at least 100%, at least 200%, at least 300%, at least 400%, at least 500%, or at least 1000%. A third boosting dose may increase the immune response by at least 50%, at least 100%, at least 200%, at least 300%, at least 400%, at least 500%, or at least 1000%.


An amount adequate to elicit an immune response is defined as a “therapeutically effective dose.” Amounts effective for this use will depend on, e.g., the composition, the manner of administration, the stage and severity of the disease being treated, the weight and general state of health of the patient, and the judgment of the prescribing physician. It should be kept in mind that immunogenic compositions can generally be employed in serious disease states, that is, life-threatening or potentially life-threatening situations, especially when the cancer has metastasized. In such cases, in view of the minimization of extraneous substances and the relative nontoxic nature of a neoantigen, it is possible and can be felt desirable by the treating physician to administer substantial excesses of these immunogenic compositions.


The immunogenic composition comprising one or more tumor-specific neoantigens can be administered to the subject alone or in combination with other therapeutic agents. The therapeutic agent can be, for example, a chemotherapeutic agent, radiation, or immunotherapy. Any suitable therapeutic treatment for a particular cancer can be administered. Exemplary chemotherapeutic agents include, but are not limited to aldesleukin, altretamine, amifostine, asparaginase, bleomycin, capecitabine, carboplatin, carmustine, cladribine, cisapride, cisplatin, cyclophosphamide, cytarabine, dacarbazine (DTIC), dactinomycin, docetaxel, doxorubicin, dronabinol, epoetin alpha, etoposide, filgrastim, fludarabine, fluorouracil, gemcitabine, granisetron, hydroxyurea, idarubicin, ifosfamide, interferon alpha, irinotecan, lansoprazole, levamisole, leucovorin, megestrol, mesna, methotrexate, metoclopramide, mitomycin, mitotane, mitoxantrone, omeprazole, ondansetron, paclitaxel (Taxol®), pilocarpine, prochloroperazine, rituximab, tamoxifen, taxol, topotecan hydrochloride, trastuzumab, vinblastine, vincristine and vinorelbine tartrate. The subject may be administered a small molecule, or targeted therapy (e.g. kinase inhibitor). The subject may be further administered an anti-CTLA antibody or anti-PD-1 antibody or anti-PD-Ll antibody. Blockade of CTLA-4 or PD-L1 by antibodies can enhance the immune response to cancerous cells in the patient.


III. Immunogenic Compositions


The invention further relates to personalized (i.e., subject-specific) immunogenic compositions (e.g., a cancer vaccine) comprising one or more tumor-specific antigens selected using the methods described herein. Such immunogenic compositions can be formulated according to standard procedures in the art. The immunogenic composition is capable of raising a specific immune response.


The immunogenic composition can be formulated so that the selection and number of tumor-specific neoantigens is tailored to the subject's particular cancer. For example, the selection of the tumor-specific neoantigens can be dependent on the specific type of cancer, the status of the cancer, the immune status of the subject, and the MHC-type of the subject.


The immunogenic composition can comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 37, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 or more tumor-specific neoantigens. The immunogenic composition can contain about 10-20 tumor-specific neoantigens, about 10-30 tumor-specific neoantigens, about 10-40 tumor-specific neoantigens, about 10-50 tumor-specific neoantigens, about 10-60 tumor-specific neoantigens, about 10-70 tumor-specific neoantigens, about 10-80 tumor-specific neoantigens, about 10-90 tumor-specific neoantigens, or about 10-100 tumor-specific neoantigens. Preferably, the immunogenic composition comprises at least about 10 tumor-specific neoantigens. Also preferably is an immunogenic composition that comprises at least about 20 tumor-specific neoantigens.


The immunogenic composition can further comprise natural or synthetic antigens. The natural or synthetic antigens can increase the immune response. Exemplary natural or synthetic antigens include, but are not limited to, pan-DR epitope (PADRE) and tetanus toxin antigen.


The immunogenic composition can be in any form, for example a synthetic long peptide, RNA, DNA, a cell, a dendritic cell, a nucleotide sequence, a polypeptide sequence, a plasmid, or a vector.


Tumor-specific neoantigens can also be included in viral vector-based vaccine platforms, such as vaccinia, fowlpox, self-replicating alphavims, marabavirus, adenovirus (See, e.g., Tatsis et al., Molecular Therapy, 10:616-629 (2004)), or lentivirus, including but not limited to second, third or hybrid second/third generation lentivirus and recombinant lentivirus of any generation designed to target specific cell types or receptors (See, e.g., Hu et al., Immunol Rev., 239(1): 45-61 (2011), Sakma et al, Biochem J., 443(3):603-18 (2012)). Dependent on the packaging capacity of the above-mentioned viral vector-based vaccine platforms, this approach can deliver one or more nucleotide sequences that encode one or more tumor-specific neoantigen peptides. The sequences may be flanked by non-mutated sequences, may be separated by linkers or may be preceded with one or more sequences targeting a subcellular compartment (See, e.g., Gros et al., Nat Med., 22 (4):433-8 (2016), Stronen et al., Science., 352(6291): 1337-1341 (2016), Lu et al., Clin Cancer Res., 20(13):3401-3410 (2014)). Upon introduction into a host, infected cells express the one or more tumor-specific neoantigens, and thereby elicit a host immune (e.g., CD8+ or CD4+) response against the one or more tumor-specific neoantigens. Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848. Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover et al. (Nature 351:456-460 (1991)). A wide variety of other vaccine vectors useful for therapeutic administration or immunization of neoantigens that will be apparent to those skilled in the art from the description herein may also be used.


The immunogenic composition can contain individualized components, according to their personal needs of the particular subject.


The immunogenic composition described herein can further comprise an adjuvant. Adjuvants are any substance whose admixture into an immunogenic composition increases, or otherwise enhances and/or boosts, the immune response to a tumor-specific neoantigen, but when the substance is administered alone does not generate an immune response to a tumor-specific neoantigen. The adjuvant preferably generates an immune response to the neoantigen and does not produce an allergy or other adverse reaction. It is contemplated herein that the immunogenic composition can be administered before, together, concomitantly with, or after administration of the immunogenic composition.


Adjuvants can enhance an immune response by several mechanisms including, e.g., lymphocyte recruitment, stimulation of B and/or T cells, and stimulation of macrophages. When an immunogenic composition of the invention comprises adjuvants or is administered together with one or more adjuvants, the adjuvants that can be used include, but are not limited to, mineral salt adjuvants or mineral salt gel adjuvants, particulate adjuvants, microparticulate adjuvants, mucosal adjuvants, and immunostimulatory adjuvants. Examples of adjuvants include, but are not limited to, aluminum salts (alum) (such as aluminum hydroxide, aluminum phosphate, and aluminum sulfate), 3 De-O-acylated monophosphoryl lipid A (MPL) (see, GB 2220211), MF59 (Novartis), AS03 (Glaxo SmithKline), AS04 (Glaxo SmithKline), polysorbate 80 (Tween 80; ICL Americas, Inc.), imidazopyridine compounds (see, International Application No. PCT/US2007/064857, published as International Publication No. WO2007/109812), imidazoquinoxaline compounds (see, International Application No. PCT/US2007/064858, published as International Publication No. WO2007/109813) and saponins, such as QS21 (see, Kensil et al, in Vaccine Design: The Subunit and Adjuvant Approach (eds. Powell & Newman, Plenum Press, NY, 1995); U.S. Pat. No. 5,057,540). In some embodiments, the adjuvant is Freund's adjuvant (complete or incomplete). Other adjuvants are oil in water emulsions (such as squalene or peanut oil), optionally in combination with immune stimulants, such as monophosphoryl lipid A (see, Stoute et al, N. Engl. J. Med. 336, 86-91 (1997)).


CpG immunostimulatory oligonucleotides have also been reported to enhance the effects of adjuvants in a vaccine setting. Other TLR binding molecules such as RNA binding TLR 7, TLR 8 and/or TLR 9 may also be used.


Other examples of useful adjuvants include, but are not limited to, chemically modified CpGs (e.g. CpR, Idera), Poly(I:C)(e.g. polyi:Cl2U), poly ICLC, non-CpG bacterial DNA or RNA as well as immunoactive small molecules and antibodies such as cyclophosphamide, sunitmib, bevacizumab, Celebrex (celecoxib), NCX-4016, sildenafil, tadalafil, vardenafil, sorafinib, XL-999, CP-547632, pazopamb, ZD2171, AZD2171, ipilimumab, tremelimumab, and SC58175, which may act therapeutically and/or as an adjuvant. In embodiments, Poly ICLC is a preferable adjuvant.


The immunogenic compositions can comprise one or more tumor-specific neoantigens described herein alone or together with a pharmaceutically acceptable carrier. Suspensions or dispersions of one or more tumor-specific neoantigens, especially isotonic aqueous suspensions, dispersions, or ampgipgilic solvents can be used. The immunogenic compositions may be sterilized and/or may comprise excipients, e.g., preservatives, stabilizers, wetting agents and/or emulsifiers, solubilizers, salts for regulating osmotic pressure and/or buffers and are prepared in a manner known per se, for example by means of conventional dispersing and suspending processes. In certain embodiments, such dispersions or suspensions may comprise viscosity-regulating agents. The suspensions or dispersions are kept at temperatures around 2° C. to 8° C., or preferentially for longer storage may be frozen and then thawed shortly before use. For injection, the vaccine or immunogenic preparations may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. The solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.


In certain embodiments, the compositions described herein additionally comprise a preservative, e.g., the mercury derivative thimerosal. In a specific embodiment, the pharmaceutical compositions described herein comprise 0.001% to 0.01% thimerosal. In other embodiments, the pharmaceutical compositions described herein do not comprise a preservative.


An excipient can be present independently of an adjuvant. The function of an excipient can be, for example, to increase the molecular weight of the immunogenic composition, to increase activity or immunogenicity, to confer stability, to increase the biological activity, or to increase serum-half life. An excipient can also be used to aid presentation of the one or more tumor-specific neoantigens to T-cells (e.g., CD 4+ or CD8+ T-cells). The excipient can be a carrier protein such as, but not limited to, keyhole limpet hemocyanin, serum proteins such as transferrin, bovine serum albumin, human serum albumin, thyroglobulin or ovalbumin, immunoglobulins, or hormones, such as insulin or palmitic acid. For immunization of humans, the carrier is generally a physiologically acceptable carrier acceptable to humans and safe. Alternatively, the carrier can be dextran, for example sepharose.


Cytotoxic T-cells recognizes an antigen in the form of a peptide bound to an MHC molecule, rather than the intact foreign antigen itself. The MHC molecule itself is located at the cell surface of an antigen presenting cell. Thus, an activation of cytotoxic T-cells is possible if a trimeric complex of peptide antigen, MHC molecule, and antigen-presenting cell (APC) is present. It may enhance the immune response if not only the one or more tumor-specific antigens are used for activation of cytotoxic T-cells, but if additional APCs with the respective MHC molecule are added. Therefore, in some embodiments an immunogenic composition additionally contains at least one APC.


The immunogenic composition can comprise an acceptable carrier (e.g., an aqueous carrier). A variety of aqueous carriers can be used, e.g., water, buffered water, 0.9% saline, 0.3% glycine, hyaluronic acid and the like. These compositions can be sterilized by conventional, well known sterilization techniques, or can be sterile filtered. The resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.


Neoantigens can also be administered via liposomes, which target them to a particular cell tissue, such as lymphoid tissue. Liposomes are also useful in increasing half-life. Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. In these preparations the neoantigen to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to, e.g., a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the CD45 antigen, or with other therapeutic or immunogenic compositions. Thus, liposomes filled with a desired neoantigen can be directed to the site of lymphoid cells, where the liposomes then deliver the selected immunogenic compositions. Liposomes can be formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, e.g., liposome size, acid lability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., An. Rev. Biophys. Bioeng. 9;467 (1980), U.S. Pat. Nos. 4,235,871, 4,501,728, 4,501,728, 4,837,028, and 5,019,369.


For targeting to the immune cells, a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells. A liposome suspension can be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the peptide being delivered, and the stage of the disease being treated.


An alternative method for targeting immune cells, components of the immunogenic composition, such as an antigen (i.e., tumor-specific neoantigen), ligand, or adjuvant (e.g., TLR) can be incorporated into an poly(lactic-co-glycolic) microspheres. The poly(lactic-co-glycolic) microspheres can entrap components of the immunogenic composition as an endosomal delivery device.


For therapeutic or immunization purposes, nucleic acids encoding a tumor-specific neoantigen described herein can also be administered to the patient. A number of methods are conveniently used to deliver the nucleic acids to the patient. For instance, the nucleic acid can be delivered directly, as “naked DNA”. This approach is described, for instance, in Wolff et al., Science 247: 1465-1468 (1990), as well as U.S. Pat. Nos. 5,580,859 and 5,589,466. The nucleic acids can also be administered using ballistic delivery as described, for instance, in U.S. Pat. No. 5,204,253. Particles comprised solely of DNA can be administered. Alternatively, DNA can be adhered to particles, such as gold particles. Approaches for delivering nucleic acid sequences can include viral vectors, mRNA vectors, and DNA vectors with or without electroporation. The nucleic acids can also be delivered complexed to cationic compounds, such as cationic lipids.


The immunogenic compositions provided herein can be administered to the subject by, including but not limited to, oral, intradermal, intratumoral, intramuscular, intraperitoneal, intravenous, topical, subcutaneous, percutaneous, intranasal and inhalation routes, and via scarification (scratching through the top layers of skin, e.g., using a bifurcated needle). The immunogenic composition can be administered at the tumor site to induce a local immune response to the tumor.


The dosage of the one or more tumor-specific neoantigens may depend upon the type of composition and upon the subject's age, weight, body surface area, individual condition, the individual pharmacokinetic data, and the mode of administration.


Also disclosed herein is a method of manufacturing an immunogenic composition comprising one or more tumor-specific neoantigens selected by performing the steps of the methods disclosed herein. An immunogenic composition as described herein can be manufactured using methods known in the art. For example, a method of producing a tumor-specific neoantigen or a vector (e.g., a vector including at least one sequence encoding one or more tumor-specific neoantigens) disclosed herein can include culturing a host cell under conditions suitable for expressing the neoantigen or vector, wherein the host cell comprises at least one polynucleotide encoding the neoantigen or vector, and purifying the neoantigen or vector. Standard purification methods include chromatographic techniques, electrophoretic, immunological, precipitation, dialysis, filtration, concentration, and chromatofocusing techniques.


Host cells can include a Chinese Hamster Ovary (CHO) cell, NS0 cell, yeast, or a HEK293 cell. Host cells can be transformed with one or more polynucleotides comprising at least one nucleic acid sequence that encodes one or more tumor-specific neoantigens or vector disclosed herein. In certain embodiments the isolated polynucleotide can be cDNA.


IV. Samples


The methods disclosed herein comprise selecting one or more tumor-specific neoantigens derived from a tumor. The methods of selecting one or more tumor-specific neoantigens comprise obtaining sequence data derived from the tumor. Such sequence data can be derived from a tumor sample of a subject. The tumor sample can be obtained from a tumor biopsy.


The tumor sample can be obtained from human or non-human subjects. Preferentially, the tumor sample is obtained from a human. The tumor sample can be obtained from a variety of biological sources that comprise cancerous tumors. The tumor can be from a tumor site or circulating tumor cells from blood. Exemplary samples can include, but are not limited to, bodily fluid, tissue biopsies, blood samples, serum plasma, stool, skin samples, and the like. The source of a sample can be a solid tissue sample such as a tumor tissue biopsy. Tissue biopsy samples may be biopsies from, e.g., lung, prostate, colon, skin, breast tissue, or lymph nodes. Samples can also be e.g., samples of bone marrow, including bone marrow aspirate and bone marrow biopsies. Samples can also be liquid biopsies, e.g., circulating tumor cells, cell-free circulating tumor DNA, or exosomes. Blood samples can be whole blood, partially purified blood, or a fraction of whole or partially purified blood, such as peripheral blood mononucleated cells (PBMCs).


The tumor samples described herein can be obtained directly from a subject, derived from a subject, or derived from samples obtained from a subject, such as cultured cells derived from a biological fluid or tissue sample. The tumor biopsy can be a fresh sample. The fresh sample can be fixed after removal from the subject with any known fixatives (e.g. formalin, Zenker's fixative, or B-5 fixative). The tumor biopsy can also be archived samples, such as frozen samples, cryopreserved samples, of cells obtained directly from a subject or of cells derived from cells obtained from a subject. Preferably, the tumor sample obtained from a subject is a fresh tumor biopsy.


The tumor sample can be obtained from a subject by any means including, but not limited to, tumor biopsy, needle aspirate, scraping, surgical excision, surgical incision, venipuncture, or other means known in the art. A tumor biopsy is a preferred method for obtaining the tumor. The tumor biopsy can be obtained from any cancerous site, for example, a primary tumor or a secondary tumor. A tumor biopsy from a primary tumor is generally preferred. Those skilled in the art will recognize other suitable techniques for obtaining tumor samples.


The tumor sample can be obtained from the subject in a single procedure. The tumor sample can be obtained from the subject repeatedly over a period of time. For example, the tumor sample may be obtained once a day, once a week, monthly, biannually, or annually. Obtaining numerous samples over a period of time can be useful to identify and select new tumor-specific neoantigens. The tumor sample can be obtained from the same tumor or different tumors.


The tumor sample can be obtained from the primary tumor, one or more metastases, and/or individual sites of tumor growth (e.g., bone marrow from different skeletal parts, such as hip, bone, or vertebra). The tumor sample can be obtained from the same site or different site.


5. EQUIVALENTS

It will be readily apparent to those skilled in the art that other suitable modifications and adaptions of the methods of the invention described herein are obvious and may be made using suitable equivalents without departing from the scope of the disclosure or the embodiments. Having now described certain compositions and methods in detail, the same will be more clearly understood by reference to the following examples, which are introduced for illustration only and not intended to be limiting.


6. EXAMPLES

The following are examples of methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided herein.


Example 1. Neoantigen Peptide Selection

This example describes the individual procedural steps to select neo-antigenic, immunogenic peptides identified from next generation sequencing data generated from a patient's tumor and normal tissue.


1.1. Sample Preparation and Generation of WES, WGS, and RNA-Seq Data

Tumor biopsies or surgical explants were collected from study participants with informed consent and transported to the Clinical Trials CLIA laboratory in tissue culture medium on ice. There, samples were accessioned, and assigned with a specific unique sample identifier. Next, tissue was weighed, portioned, and placed in five (5) times its volume of RNAlater Stabilization Solution (ThermoFisher, CatNo AM7020). The samples were then left overnight at 4° C., removed from the RNAlater solution, and placed in a cryovial with 1 mL STEMCELL CroyStor10 (CatNo 07952) and transferred into a CoolCell (Corning, CatNo 432000) at −80° C.


Peripheral Blood collected in ACD tubes from participants in the accompanying Study Protocol, were transported to a Specimen Processing & Research Cell Bank, where PBMC processing occured according to SOP using Ficoll. PBMC processing may occur prior to tumor biopsy to allow simultaneous shipping of PBMC and tumor biopsy tissues to a sequencing provider.









TABLE 1







Samples














Tumor
Manufacturing Steps Performed















Sample


Sample


RNA
HLA
Peptide


ID
Biopsy Type
Gender
Type
WES
WGS
Seq
Haplotyping
Manufacturing





AAAAA
Subcutaneous
Male
Single cell
X

X
X




metastasis

suspension1







BBBBB
Lymph node
Female
Single cell
X

X
X




metastasis

suspension1







CCCCC
Lymph node
Male
Single cell
X

X
X




metastasis

suspension1







DDDDD
Lymph node
Male
Single cell
X

X
X




metastasis

suspension1







EEEEE
Lymph node
Male
Mock
X
X
X
X
X



metastasis

biopsy










from tumor










tissue










(156 mg










tissue)2







FFFFF
Lymph node
Male
Single cell
X
X
X
X
X



metastasis

suspension










(100,000










cells)1







GGGG
Lymph node
Female
Mock
X
X
X
X
X



metastasis

biopsy










from tumor










tissue (30










mg tissue)3





Notes:



1Single cell suspensions were generated through tissue digest and disruption




2Two passes with a 3 mm punch tool




3A total of 10 passes with 14 gauge needle







All specimens (tumor biopsy and PBMC) were sent by the CLIA laboratory overnight to the sequencing provide on dry ice.


DNA, RNA, and miRNA were simultaneously isolated from the same tissue or cell specimen using the AllPrep DNA/RNA/miRNA Universal Kit (QIAGEN).


DNA/RNA sample quality and quantity were assessed using adequeate methods (e.g. Qubit, BioAnalyzer) and the following metrics were recorded:


DNA: concentration (ng/μL), total amount (ng), volume (μL)


RNA: concentration (ng/μL), total amount (ng), volume (μL), purity (RIN)









TABLE 2







Genomic DNA and Total RNA Yields from Engineering Run Samples RNA


















Amount
Concentration
Volume
Amount
Concentration

Amount
Concentration
Volume




(ng)
(ng/mL)
(μL)
(ng)
(ng/nL)
Volume
(ng)
(ng/nL)
(μL)
RIN





AAAAA
   11
  1
16
 341
34
10
  171
 24
 8
7.2





BBBBB
  126
  8
16
 239
24
10
  172
 25
 8
3.3





CCCCC
   12
  1
16
 180
18
10
  173
 25
 8
4.5





DDDDD
   83
  5
16
 160
16
10
  195
 24
 8
2.6





EEEEE
23144
263
88
4039
46
88
40035
702
57
6.8





FFFFF
 4136
 47
88
4391
50
88
 1427
 25
57
8.4





GGGGG
  203
  2
88
3467
39
88
44433
780
57
5.4









DNA samples containing more than 200 ng of genomic DNA were aliquoted, with 200 ng used for WES, and remaining DNA was shipped for another sequencing provider for WGS.


NGS was performed. An overview of the library preparation and sequencing strategies is show in Table 3 below.









TABLE 3







Details of Next Generation Sequencing performed in


PNV-21 manufacturing












NGS

Sequencing
Sequencing



Method
Target Depth
Strategy
Instrument







WES
 75× Normal,
100 bp PE
NovaSeq 6000




125× Tumor





RNA-Seq
100M
 50 bp PE
Nova Seq 6000



WGS
250× Normal,
101 bpPE
Nova Seq 6000




250× Tumor










WES data from paired tumor/normal samples was used to identify germline and somatic variants of the patient and her/his tumor.


WES is performed at a commercial sequencing vendor using the Agilent SureSelect All Exon v6 bait kit and libraries were generated and sequenced on an Illumina NovaSeq6000 instrument. DNA from PBMC samples underwent WES using a 100bp PE strategy, 75× average coverage, and a target of 38 million reads. DNA from tissue samples undergoes WES using a 100 bp PE strategy, 125× average coverage, and a target of 63 million reads.


RNA sequencing data was used to identify neo-antigen encoding RNA transcripts with sufficiently high expression, as well as to independently confirm WES derived somatic variants.


RNA was sequenced using the Illumina Stranded mRNA sequencing method using a 50bp PE strategy and a target of 100 million reads.


Sequencing libraries were created using the Illumina TruSeq Stranded mRNA method, preferentially selecting for messenger RNA by taking advantage of the polyadenylated tail.


WGS data was used to perform CNV calling and to identify subclones of the tumor samples.


WGS was performed at a commercial, CLIA validated laboratory from tumor and normal genomic DNA prepared as detailed above. Two pooled libraries from the same individual were sequenced on an Illumina S4 flow cell (FC) with read length of 2×101. Data generated on an FC with Q30>80% and error rate <3% was passed for demultiplexing.


NGS data in FASTQ format was transferred from the sequencing vendors for further bioinformatics analysis.


HLA typing was performed with a molecular assay at an accredited clinical immunogenetics laboratory.


1.2. Bioinformatics Analysis of NGS Data

Mapping and Alignment of NGS reads to the hg19 reference genome was done using the Illumina DRAGEN Bio-IT Platform (v3.8.5). Details about the input and output files, as well as the automated execution of the processing steps used for mapping and alignment are described in FIG. 2


All NGS analyses were performed using the hg19 human reference genome assembly (initial UCSC reference assembly, based on initial GRCh37 release, md5 checksum a244d8a32473650b25c6e8e1654387d6, downloaded from the Sentieon reference bundle. Mapping between known ENSEMBL genes and hg19 chromosomal coordinates (GTF file) was downloaded from UCSC. This file is used for quantification of RNA gene expression. The DRAGEN Bio-IT platform was used to generate a series of hash-table files, which are needed for mapping, alignment, and variant calling. The generated hash table files and the hg19 ENSEMBLE GTF file were uploaded to an S3 data storage bucket.


The DNA sequence mapping and alignment step took NGS FASTQ files as input and aligns reads to the provided reference genome hash tables, independently for normal and tumor (if provided) samples. Normal mapping/alignment included the generation of germline variant calls, used in Module A3 for somatic CNV calling (termed B-Allele file).









TABLE 4





NGS FASTQ input

















Module A1
DNA Seq Mapping/Alignment--




NORMAL









Environment
Version


Illumina DRA GEN Bio-IT Platform
3.8.5







Command Line


dragen -f -r $BUNDLE_FOLDER −1 $NORMAL_FASTQ 1 −2 $NORMAL_FASTQ2 \


 --RGID “${NORMAL_GROUP}” --RGSM “${NORMAL_SAMPLE}” \


 --RGPL ILLUMINA \


 --dupmark-version sort \


 --enable-variant-caller true \


 --enable-map-align-output true \


 --enable-vcf-compression true \


 --intermediate-results-dir ${TMP_FOLDER} \


 --enable-bam-indexing true \


 --output-directory ${ANALYSIS_FOLDER}/ \


 --output-file-prefix ${SM_Prefix} \


 --lic-server ${LICENSE_STRING}





Input









Variable
Value
Description





-f

Force overwrite


-r
$BUNDLE_FOLDER
Location of Hash Table folder


−1, −2
$NORMAL_FASTQ1,
Location of normal FASTQ files



$NORMAL_FASTQ2



--RGID
${NORMAL_GROUP}
Read group ID string, normal


--RGSM
${NORMAL_SAMPLE}
Read group Sample Name string, normal


--RGPL
ILLUMINA
Read group platform string, normal


--dupmark-version
sort
Enable memory optimized germline calling


--enable-variant-caller
true
Enable variant calling


--enable-map-align-
true
Output .BAM file


output




--enable-vcf-
true
Enable vcf compression


compression




--intermediate-results-
${TMP_FOLDER}
Temporary files directory, scratch


dir




--enable-bam-
true
Enable indexing of BAM files


indexing




--output-directory
${ANALYSIS_FOLDER}
Local directory for file output


--output-file-prefix
${SM_Prefix}
Prefix for output files, sample name


--lic-server
${LICENSE_STRING}
String with




User:Password@license.edicogenome.com










Output





A number of files were generated inside the output directory, all prefixed with ${SM_Prefix}:


 • Normal BAM file (plus index)


 • Normal VCF file (plus index), unfiltered and “hard-filtered”


 • Mapping quality metrics report


 • FastQC report









In addition to the mapping/alignment algorithm, this module utilized DRAGEN's reporting function (including mapping statistics and trimming report), which was used for quality control.


For tumor sample mapping/alignment, the command line was outlined in Table 5.









TABLE 5





Comand line for tumor sample mapping/alignment

















Module A1
DNA Seq Mapping/Alignment--




TUMOR









Environment
Version


Illumina DRA GEN Bio-IT Platform
3.8.5







Command Line


dragen -f-r $BUNDLE_FOLDER −1 $TUMOR_FASTQ1 −2 $TUMOR_FASTQ2 \


 --enable-variant-caller false \


 --RGID “${TUMOR_GROUP}” --RGSM “${TUMOR_SAMPLE}” \


 --RGPL ILLUMINA \


 --dupmark-version sort \


 --enable-map-align-output true \


 --enable-vcf-compression true \


 --intermediate-results-dir ${TMP_FOLDER} \


 --enable-bam-indexing true \


 --output-directory ${ANALYSIS_FOLDER}/ \


 --output-file-prefix ${SM_Prefix} tumor \


 --lic-server ${LICENSE_STRING}





Input









Variable
Value
Description





-f

Force overwrite


-r
SBUNDLE_FOLDER
Location of Hash Table folder


−1, −2
STUMOR_FASTQ1,
Location of tumor FASTQ files



STUMOR_FASTQ2



--RGID
${TUMOR_GROUP}
Read group ID string, normal


--RGSM
${TUMOR_SAMPLE}
Read group Sample Name string, normal


--RGPL
ILLUMINA
Read group platform string, normal


--enable-variant-caller
true
Enable variant calling


--dupmark-version
sort
Enable memory optimized germline calling


--enable-map-align-
true
Output. BAM file


output




--enable-vcf-
true
Enable vcf compression


compression




--intermediate-results-
${TMP_FOLDER}
Temporary files directory, scratch


dir




--enable-bam-
true
Enable indexing of BAM files


indexing




--output-directory
${ANALYSIS_FOLDER}
Local directory for file output


--output-file-prefix
${SM_Prefix}
Prefix for output files, sample name


--lic-server
${LICENSE_STRING}
String with




User:Password@license.edicogenome.com










Output





A number of files were generated inside the output directory, all prefixed with ${SM_Prefix}:


 • Tumor BAM file (plus index)


 • Mapping quality metrics report


 • FastQC report









RNA Seq FASTQ files were aligned to the UCSC hg19 human reference genome using the DRAGEN Bio-IT Platform (v3.8.5) RNA module with quantification and gene fusion detection enabled. A GENCODE hg19/GRCg37.p13 GTF file was used to map genes and gene transcripts (Ensembl gene and transcript IDs) to genomic regions.









TABLE 6





Comand line for RNA Sequence Alignment and RNA Quantification

















Module A2
RNA Seq Alignment and RNA Quantification









Environment
Version


Illumina DRA GEN Bio-IT Platform
3.8.5







Command Line


dragen -f -r $BUNDLE_FOLDER \


 -a ${USE_BUNDLE_GFF} \


 −1 $RNA_FASTQ1 −2 $RNA_FASTQ2 \


 --enable-map-align-output true \


 --enable-rna true V


 --enable-rna-quantification true \


 --enable-rna-gene-fusion true \


 --enable-bam-indexing true \


 --enable-variant-caller false \


 --RGID “${RNA_GROUP}” \


 --RGSM “${RNA_SAMPLE}” \


 --output-directory ${ANALYSIS_FOLDER}/ \


 --output-file-prefix ${SM_Prefix} RNA \


 --intermediate-results-dir ${IMP_FOLDER} \


 --lic-server ${LICENSE_STRING}





Input









Variable
Value
Description





-f

Force overwrite


-r
$BUNDLE_FOLDER
Location of Hash Table folder


-a
${USE_BUNDLE_GFF}
Location of .GFF/GTF file


−1, −2
$RNA_FASTQ1,
Location of RNA Seq FASTQ files



$RNA_FASTQ2



--enable-map-align-output
true
Enable mapping/alignment


--enable-rna
true
Enable RNA processing


--enable-rna-quantification
true
Enable quantification of gene expression


--enable-rna-gene-fusion
true
Enable detection of gene fusions


--enable-bam-indexing
true
Enable indexing of .BAM file


--enable-variant-caller
False
Disable Variant Calling


--RGID
${RNA_GROUP}
Read Group ID


--RGSM
${RNA_SAMPLE}
Read Group Sample Name


--output-directory
${ANALYSIS_FOLDER}/
Output folder for result files


--output-file-prefix
${SM_Prefix}_RNA
Prefix for output files, SM_Prefix is sample




name


--intermediate-re suits-dir
${TMP_FOLDER}
Temporary files directory, scratch


--lic-server
${LICENSE_STRING}
String with




User:Password@license.edicogenome.com










Output





A number of files were generated inside the output directory, all prefixed with ${SM_Prefix}_RNA:


 • RNA BAM file


 • RNA gene expression and transcript quantification files ${SM_Prefix}_RNA. quant.genes.sf and


   ${SM Prefix} RNA.quant.sf, respectively.


 • read trimming report









Normal (PBMC) WES and WGS.BAM files, aligned to the UCSC hg19 human reference genome, were used individually to generate a list (.VCF file) of germline variant calls, indicating differences in the study participant's genome versus the reference genome. Variants detected were single or multiple base mutations, insertions, and deletions. Structural variants were not processed. The detailed command line argument to derive germline variant calls and produce a non-filtered, and filtered germline .VCF file with the DRAGEN Bio-IT Platform (v3.8.5) is described below.


The resultant germline .VCF file was used for ensuring that candidate vaccine peptides are not a representation of the study participant's germline sequence (self-peptides), and further serves CNV calling as input B-allele frequency file.


Somatic variant occurring as differences between tumor and normal study participant samples were identified from WES and WGS aligned .BAM files using the DRAGEN Bio-IT Platform. In the first step, tumor and normal .BAM files were compared to identify tumor specific (somatic) DNA mutations, which were output as .VCF files, one non-filtered, and one filtered for high confidence variants. In a second step, and for WGS only, CNV calls were produced and output as .VCF file. Input for this step were tumor and normal .BAM files, as well as the hard-filtered germline variant call file, used as B-allele frequency input.


Additional details and command line interface options for executing the somatic variant and CNV calling are described in Table 8.









TABLE 7





Automated workflow for DNA somatic variant calling
















Module A3
DNA Somatic Variant Calling








Environment
Version


Illumina DRA GEN Bio-IT Platform
3.8.5







Command Line


 dragen -v \


  -f\


  -r $BUNDLE_FOLDER \


  --intermediate-results-dir $TMP_FOLDER \


  --enable-cnv false \


  --enable-map-align false \


  --enable-variant-caller true \


  --dupmark-version sort \


  --enable-vcf-compression true \


  --bam-input ${normal_input} \


  --tumor-bam-input ${tumor_input} \


  --output-directory ${ANALYSIS_FOLDER}/ \


  --output-file-prefix ${SM_Prefix}-TN \


  --lic-server ${LICENSE_STRING} \


  $BED_OPTION





Input








Variable
Value












-f

Force overwrite


-r
$BUNDLE_FOLDER
Location of Hash Table folder


-v

Verbose output


--intermediate-
${TMP_FOLDER}
Temporary files directory, scratch


results-dir




--enable-cnv
false
Disable CNV calling.


--enable-map-align
false
Disable map/align step


--enable-variant-
true
Enable variant calling


caller




--dupmark-version
sort
Enable memory optimized germline calling


--enable-vcf-
true
Enable vcf compression


compression




--bam-input
${normal_input}
Normal .BAM output from Module A2


--tumor-bam-input
${tumor_input}
Tumor .BAM output from Module A2


--output-directory
${ANALYSIS_FOLDER}/
Output folder for result files


--output-file-prefix
${SM_Prefix}-TN
Prefix for output files, SM_Prefix is sample name


--lic-server
${LICENSE_STRING}
String with




User:Password@license.edicogenome.com


Optional




$BED_OPTION
-vc-target-bed ${BEDFILE}
Use a .BED file to restrict processing.










Output





A number of files were generated inside the output directory, all prefixed with ${SM_Prefix}-TN:


 • Somatic .VCF file


 • Somatic .VCF file with default filters applied, ${SM_Prefix}-TN-hard.filtered.vcf


 • CNV calling .VCF, .GFF3 files


 • Additional analytics files









The somatic .VCF file (hard-filtered) were used for downstream vaccine peptide selection modules.


CNV calling in DRAGEN requires a B-Allele .VCF file. CNV calling was preceded by germline calling and normal/tumor .BAM files from Module A1.









TABLE 8





Module A4 DNASomatic CNV Calling

















Module A3
DNA Somatic CNV Calling









Environment
Version


Illumina DRA GEN Bio-IT Platform
3.8.5







Command Line


 dragen -v \


  -f \


  -r $BUNDLE_FOLDER \


  --intermediate-results-dir $TMP_FOLDER \


  --enable-cnv true \


  --cnv-enable-plots true \


  --cnv-normal-b-allele-vcf ${USE_DRAGENgermline_hf_vcf} \


  --enable-map-align false \


  --enable-variant-caller false \


  --bam-input ${normal_input} \


  --tumor-bam-input ${tumor_input} \


  --output-directory ${ANALYSIS_FOLDER}/ \


  --output-file-prefix ${SM_Prefix}-TN \


  --lic-server ${LICENSE_STRING} \


  $BED_OPTION





Input









Variable
Value
Description





-f

Force overwrite


-r
$BUNDLE_FOLDER
Location of Hash Table folder


-v

Verbose output


--intermediate-
${TMP_FOLDER}
Temporary files directory, scratch


results-dir




--enable-cnv
true
Enable CNV calling.


--cnv-enable-
true
Enable plot output


plots




--cnv-normal-b-
${USE_DRAGENgermline_hf_vcf}
B-Allele file; Location of the normal germline


allele-vcf

VCF file (hard-filtered)


--enable-map-
false
Disable map/align step


align




--enable-variant-
false
Disable variant calling


caller




--bam-input
${normal_input}
Normal .BAM output from Module A2


--tumor-bam-
${tumor_input}
Tumor .BAM output from Module A2


input




--output-directory
${ANALYSIS_FOLDER}/
Output folder for result files


--output-file-
${SM_Prefix}-TN
Prefix for output files, SM_Prefix is sample


prefix

name


--lic-server
${LICENSE_STRING}
String with




User:Password@license.edicogenome.com


Optional




$BED_OPTION
-vc-target-bed ${BEDFILE}
Use a .BED file to restrict processing.










Output





A number of files will be generated inside the output directory, all prefixed with ${SM_Prefix}-TN:


 • CNV calling .VCF, .GFF3 files


 • Additional analytics files









In order to analyze mapping/alignment efficacy across contigs, Module A-QC1 employed the mosdepth analysis program taking a .BAM file as input. For WES, additionally, a .BED file to restrict analysis to defined genomic regions was provided. The hg19 bed file for the Agilent Sure Select A11 Exon v6 capture bait set was download from the Agilent website and stored in S3 for automated download by the processing pipeline.









TABLE 9





Module A-QC1 Alignment Coverage Analysis

















Module A-QC1
Alignment Coverage Analysis









Environment
Version


mosdepth (see: https://github.com/btentp/mosdepth)
0.3.1







Command Line


mosdepth -t 4 -n -x -q 1000 ${mosdepth_bedoption} ${ANALYSIS_FOLDER}/S{SM_Prefix}_tumor


$USE_TUMOR_BAM





Input









Variable
Value
Description





Positional 1
${ANALYSIS_FOLDER}/${SM_Prefix} tumor
Output file location




and prefix (here shown




for tumor BAM




analysis)




ANALYSIS FOLDER




is the local output




folder




SM_prefix is the




sample name prefix


Positional 2
$USE_TUMOR_BAM
Input BAM file (here




shown for tumor BAM




analysis)


-t
  4
Number of threads


-n

Do not output per base




analysis


-x

Fast mode


-q
1000
Write quantized output


Optional:




mosdepth_bedoption
-b ${BEDFILE}
Use BED file to restrict




analysis (WES only).










Output





A number of files were generated inside the output directory, all prefixed with ${SM_Prefix}_tumor


(/or_normal):


 • Summary file


 • Global distribution file


Additional output, see mosdepth manual (https://github.com/brentp/mosdepth)









The mosdepth summary file is a tab delimited text file which indicates the mean coverage (column mean) for all contigs present in the .BAM file (column chrom). If a .BED file was provide, the suffix “_region” denotes metrics for a .BED restricted contig. An abbreviated listing of a mosdepth summary file is shown (e.g., Table 10).









TABLE 10







Mosdepth Summary File












chrom
length
bases
mean
min
max















chrM
16571
385781
23.28
7
91


chrM_region
0
0
0.00
0
0


chr1
249250621
974106896
3.91
0
4709


chr1_region
6066056
716175291
118.06
0
4709


chr2
243199373
680541943
2.80
0
3488


chr2_region
4449345
484022026
108.79
0
1162


chr3
198022430
529198467
2.67
0
2197


chr3_region
3462072
381197919
110.11
0
2197









For analysis and quality control, the tumor and normal mosdepth summary files wereused to generate specification reports.


Example Results from Mapping Aligning are shown in Tables 11 to 15 below.









TABLE 11







WES normal (PBMC) sample per contig and total alignment coverage


Normal WES per contig coverage














contig
AAAAA
BBBBB
CCCCC
DDDDD
EEEEE
FFFFF
GGGGG

















chr1
91.04
75.46
83.98
118.06
88.65
124.08
134.29


chr2
84.07
69.79
77.71
108.79
78.85
112.96
124.92


chr3
85.35
71.39
78.97
110.11
80.91
114.52
124.62


chr4
85.64
73.11
80.79
114.08
80.04
113.62
131.06


chr5
85.44
70.48
78.06
110.34
79.30
113.73
124.23


chr6
87.31
71.82
79.40
110.67
80.76
116.11
125.74


chr7
90.42
75.10
82.71
117.22
87.48
124.19
132.08


chr8
94.21
75.16
85.69
123.53
89.09
128.33
137.32


chr9
90.26
75.37
82.87
117.47
87.68
125.38
133.51


chr10
84.24
69.72
76.61
107.61
79.14
112.51
123.26


chr11
93.49
77.96
85.98
119.83
90.87
127.68
135.81


chr12
86.84
71.41
79.31
110.07
80.74
115.08
125.06


chr13
81.67
66.81
73.89
103.30
73.50
105.97
119.95


chr14
87.89
73.11
81.47
114.51
83.61
119.02
128.97


chr15
87.06
75.04
81.88
115.35
88.59
123.23
130.83


chr16
104.70
88.37
96.08
138.82
106.57
150.20
156.55


chr17
98.46
82.36
90.71
126.92
97.05
135.76
142.66


chr18
81.52
66.73
73.83
104.19
74.26
107.36
119.56


chr19
106.65
89.89
98.79
141.60
108.96
154.08
157.96


chr20
92.79
78.01
85.05
120.45
91.93
129.56
136.70


chr21
94.85
76.80
84.22
118.44
90.22
126.95
135.70


chr22
99.98
84.02
91.46
129.71
102.41
141.08
145.47


chrX
51.30
79.83
46.91
65.47
48.16
68.42
143.35


chrY
67.52
11.87
60.97
94.91
60.38
94.90
22.04


total
89.44
75.69
82.25
115.95
86.30
122.25
133.61
















TABLE 12







WES tumor sample per contig and total alignment coverage


Tumor WES per contig coverage














contig
AAAAA
BBBBB
CCCCC
DDDDD
EEEEE
FFFFF
GGGGG

















chr1
111.45
156.87
165.12
130.31
168.65
175.27
167.75


chr2
96.21
140.58
134.34
108.12
121.00
132.45
194.87


chr3
101.00
143.05
135.00
106.33
124.52
160.68
183.05


chr4
90.88
145.18
136.25
117.13
123.96
157.44
213.40


chr5
94.44
141.95
146.51
120.00
122.43
133.47
132.74


chr6
101.29
148.37
141.70
113.56
132.15
152.03
151.15


chr7
110.34
140.52
217.99
182.57
130.89
222.42
223.98


chr8
114.25
170.43
250.80
214.17
280.41
152.57
194.43


chr9
113.12
149.56
128.87
102.43
76.08
149.03
177.16


chr10
95.86
138.91
126.65
98.06
120.73
107.09
92.70


chr11
118.91
158.83
156.03
121.32
105.10
151.13
144.34


chr12
101.13
148.19
136.95
108.99
150.62
135.60
182.38


chr13
85.61
136.52
127.46
107.24
117.89
123.71
103.11


chr14
104.96
149.85
144.83
116.11
128.92
140.17
95.47


chr15
105.21
149.91
180.22
139.82
164.08
182.73
182.92


chr16
140.71
173.30
173.31
138.51
151.85
179.80
148.28


chr17
132.27
165.93
155.34
120.67
148.50
183.20
90.28


chr18
84.42
125.96
136.40
110.64
110.70
149.78
188.79


chr19
143.86
182.17
199.42
158.86
154.05
183.79
147.17


chr20
118.41
153.07
157.90
124.87
150.00
194.95
169.70


chr21
111.10
149.81
165.03
135.30
132.81
175.10
182.38


chr22
135.02
173.50
186.06
141.07
145.66
168.84
135.34


chrX
58.26
145.37
79.38
63.49
73.73
110.56
197.29


chrY
65.76
20.79
84.53
78.93
88.48
77.81
23.72


total
108.84
152.51
156.13
125.15
137.69
158.47
161.36
















TABLE 13







WGS normal (PBMC) sample per contig


and total alignment coverage


Normal WGS per contig coverage












contig
DDDDD
FFFFF
GGGGG
















chr1
408.85
335.84
437.51



chr2
440.92
362.63
469.03



chr3
440.34
362.47
467.50



chr4
442.12
362.43
467.41



chr5
442.23
362.20
468.21



chr6
443.38
361.16
465.83



chr7
438.07
360.01
465.85



chr8
446.34
363.57
468.54



chr9
377.33
311.09
403.67



chr10
431.20
354.94
460.37



chr11
435.27
358.04
464.53



chr12
435.37
358.91
464.05



chr13
372.30
306.15
394.59



chr14
372.68
303.29
392.87



chr15
359.90
297.52
383.09



chr16
450.19
335.70
438.14



chr17
431.80
354.46
463.96



chr18
429.83
353.34
456.54



chr19
424.91
349.23
459.91



chr20
423.16
348.67
454.41



chr21
346.86
283.98
368.69



chr22
307.28
250.94
330.83



chrX
223.25
188.30
454.42



chrY
150.58
104.17
17.89



total
407.22
331.83
438.21

















TABLE 14







WGS Tumor sample per contig


and total alignment coverage


Tumor WGS per contig coverage












contig
DDDDD
FFFFF
GGGGG
















chr1
282.48
273.39
66.93



chr2
270.31
249.52
94.40



chr3
267.23
295.22
95.49



chr4
281.26
295.03
104.84



chr5
304.11
249.11
76.37



chr6
282.90
264.26
68.60



chr7
427.86
366.02
95.56



chr8
380.36
250.46
93.94



chr9
206.64
215.87
72.53



chr10
244.67
197.83
44.53



chr11
276.04
245.76
71.68



chr12
266.30
246.75
90.22



chr13
236.75
210.90
46.36



chr14
236.57
208.52
40.28



chr15
271.98
255.83
68.02



chr16
278.66
230.60
49.83



chr17
258.87
275.75
36.02



chr18
278.98
287.59
92.71



chr19
296.80
239.13
48.70



chr20
257.91
287.12
71.35



chr21
243.95
227.28
72.47



chr22
208.05
172.38
33.75



chrX
133.49
174.43
91.72



chrY
74.88
48.51
2.64



total
270.11
249.38
74.13

















TABLE 15







RNA seq tumor sample per contig and total alignment coverage


tumor RNA per contig coverage














contig
AAAAA
BBBBB
CCCCC
DDDDD
EEEEE
FFFFF
GGGGG

















chr1
3.64
5.06
5.37
4.30
5.62
5.73
5.54


chr2
2.43
3.50
3.39
2.77
3.11
3.37
4.96


chr3
2.39
3.37
3.22
2.58
3.01
3.85
4.46


chr4
1.56
2.43
2.33
2.02
2.17
2.72
3.69


chr5
1.96
2.90
3.06
2.54
2.60
2.79
2.85


chr6
2.43
3.52
3.41
2.77
3.17
3.66
3.67


chr7
2.86
3.58
5.58
4.73
3.44
5.76
5.79


chr8
2.19
3.31
4.71
4.04
5.61
2.98
3.92


chr9
2.69
3.54
3.10
2.50
1.86
3.59
4.34


chr10
2.43
3.46
3.21
2.52
3.10
2.72
2.38


chr11
4.00
5.33
5.27
4.16
3.63
5.12
5.05


chr12
3.32
4.80
4.47
3.61
5.02
4.46
6.07


chr13
1.15
1.79
1.72
1.47
1.62
1.69
1.39


chr14
2.61
3.69
3.63
2.95
3.26
3.51
2.43


chr15
3.22
4.50
5.48
4.31
5.05
5.57
5.61


chr16
5.38
6.61
6.65
5.37
5.91
6.89
5.77


chr17
7.34
9.08
8.56
6.73
8.31
10.13
5.06


chr18
1.46
2.12
2.35
1.93
1.94
2.60
3.27


chr19
11.51
14.44
15.92
12.81
12.48
14.67
11.96


chr20
3.70
4.73
4.90
3.92
4.75
6.06
5.39


chr21
2.00
2.65
2.96
2.47
2.44
3.16
3.35


chr22
4.84
6.12
6.60
5.08
5.28
6.00
4.86


chrX
1.15
2.85
1.57
1.28
1.49
2.21
4.01


chrY
0.38
0.11
0.48
0.45
0.53
0.44
0.12


total
2.92
4.05
4.18
3.39
3.79
4.28
4.45









Results of the Variant Calling Process is shown in Tables 16-19.









TABLE 16







Number of WES normal (PMC) germline variants















Regions
metric
AAAAA
BBBBB
CCCCC
DDDDD
EVEEE
FFFFF
GGGGG


















all











number
30,973
25,934
32,148
50,833
29,681
47,556
51,418



of










indels










number
209,535
180,749
219,602
307,090
204,141
294,761
307,494



of










records










number
178,629
154,862
187,522
256,349
174,505
247,278
256,162



of










SNPs









CDS











number
1,105
1,061
1,104
1,120
1,055
1,110
1,066



of










indels










number
33,753
33,422
33,595
33,764
33,364
33,733
33,402



of










records










number
32,653
32,366
32,494
32,646
32,310
32,628
32,342



of










SNPs
















TABLE 17







Number of WES tumor/normal somatic variants















Regions
metric
AAAAA
BBBBB
CCCCC
DDDDD
EEEEE
FFFFF
GGGGG


















all











number of
23
8
14
11
33
49
21



indels










number of
288
163
605
619
305
3,329
180



records










number of
265
155
591
608
272
3,280
159



SNPs









CDS











number of
11
3
6
1
17
17
3



indels










number of
204
85
344
346
132
1,528
66



records










number of
193
82
338
345
115
1,511
63



SNPs
















TABLE 18







Number of WGS normal germline variants











Regions
metric
DDDDD
FFFFF
GGGGG














all







number of indels
931,451
934,986
939,511



number of records
4,693,517
4,705,612
4,727,192



number of SNPs
3,767,056
3,775,609
3,792,797


CDS







number of indels
1,190
1,149
1,119



number of records
34,546
34,431
34,649



number of SNPs
33,359
33,287
33,536
















TABLE 19







Number of WGS tumor/normal somatic variants











Regions
metric
DDDDD
FFFFF
GGGGG














all







number of indels
3,186
7,884
928



number of records
29,927
182,205
5,103



number of SNPs
26,741
174,321
4,175


CDS







number of indels
3
16
2



number of records
366
1,570
54



number of SNPs
363
1,554
52









The subsequent module of workflow processed tumor somatic variants and copy number variants called by Module A from tumor-normal WGS samples and output a membership probability for each somatic variant into a set of N tumor-specific sub-clones, where N is a parameter output by the module. It also output an estimate of cellular prevalence for each mutation. This Module also performed bulk deconvolution of variants into sub-clones. The WGS files were generated from DRAGEN in Module A and processed in this module to estimate sub-clonality from cellular prevalence of somatic variants. FIG. 3 shows a workflow diagram of the module for clonality deconvolution.


1.3. Peptide Selection from NGS Data


Vaccine peptides to be manufactured were selected by a peptide prediction and machine learning algorithms within a personalize peptide prediction pipeline (p4vax). All components of this software solution are briefly described herein.


Mapping/Alignment, RNA gene expression, germline variants, somatic variants, and CNV caller output files, as well as the results of HLA haplotyping were ingested by the peptide prediction workflow to:


1. Identify variants with coding effects on expressed RNA


2. Select putative MHC class I and II binding peptides spanning across the non-synonymous variant


3. Confirm presence of variant in RNA sequencing reads


4. Estimate MHC class I and II processing, presentation, and immunogenicity


5. Confirm presence of mutation only in tumor DNA but not in germline (normal) DNA by correcting hg19 reference with germline variant calls


6. Filter potentially toxic peptides or products of endo- or exo-peptidase metabolism


7. Rank peptides on MHC class I and II processing, presentation, and immunogenicity


8. Maximize the expected percent of tumor cells targeted by the peptides


Specifically, for peptide selection, the results from WES and WES based somatic variant calling were used.


Using a tumor sub-clone deconvolution algorithm, the workflow processed tumor somatic variants & copy number variants called by Module A from tumor-normal WGS samples and additionally output a membership probability for each somatic variant into a set of N tumor-specific clusters of mutation/cellular prevalence. The results from this step were used in step 8.


The output of the peptide prediction pipeline is an exhaustive list of potential vaccine peptides ranked by a combination of MHC class I and II binding/presentation scores, immunogenicity, and tumor cellular prevalence clusters.


From this ranked list of potential peptide synthesis candidates, a set of up to 80 peptides ranked highest in the list was selected. After manual verification of the existence of somatic variants, peptide sequences resulting from verified somatic variants were communicated to a peptide manufacturer for synthesis.


Peptide Selection was performed for the Work Example samples DDDDD, FFFFF, and GGGGG (Table 20-Table 22) and twenty peptides were selected further as candidates for pool formulation.









TABLE 20







Peptide Selection for engineering run sample DDDDD





















SEQ




Short/





ID
MHC I
MHC II


Lon
Rank
Gene
Variant
Sequence
Length
NO:
score
score


















long
 1
MOGS
chr2: 74690507: C: T
QDSNTSALPLVSLFF
15
 2
 0.07329569
  0.66997391



 2
EXOC7
chr17: 74079782: C: T
YIKYKVEQVGDMIDRL
16
 3
 0.05583751
  0.62314493



 3
MEGF8
chr19: 42858849: C: T
GQRRDRLLTVQALSGL
16
 4
 0.05151247
  0.73578168



 4
EXOC7
chr17: 74079781: C: T
YIKYKVEQVGDMIDRLF
17
 5
 0.05031836
  0.63131081



 5
LIMA1
chr12: 50571454: G: A
EGIKMSKPKWPLEDEISKPE
22
 6
 0.20668915
  0.43862386






VP







 6
PTPN4
chr2: 120714433: A: C
QFDQLYRTKPGMTMSC
16
 7
 0.03901816
  0.65768236



 7
BRAF
chr7: 140414433: A: C
GDVAVKMLTPQQLQA
15
 8
0.0365321
  0.67463777





TAGGTGCTGTCACAT










(SEQ ID NO: 1): G








 8
GINS4
chr8: 41393899: T: A
RLEQAWMNEKLAPELLESKP
20
 9
 0.03424379
  0.51320203



 9
GAS8
chr16: 90106895: C: G
NEVLAASNLDPAALRLVSRK
22
10
 0.02816643
  0.55796491






LE







10
PRKCZ
chr1: 2066754: G: A
GARRWRKLYRANSHLFQAKR
22
11
0.0278652
  0.57790382






FN







11
RPS6KB2
chr11: 67200282: C: T
SQGIIYRDLKLENIMLSSQG
22
12
 0.13305076
  0.58418771






HI







12
BST1
chr4: 15716974: C: T
GSEPTGAYSIKGFFADYEIP
21
13
 0.02372363
  0.73712491






N







13
HIPK2
chr7: 139316445: G: A
YRAPEIILGLLFCEAI
16
14
 0.02345472
  0.78064901



14
SYNJ1
chr21: 34017988: G: A 
APPTRLAPPQRPPPPS
16
15
 0.01945223
  0.46290062



15
EPS8L3
chr1: 110293322: C: T
LQMLCPQEAPQILSRLEAVR
21
16
 0.09542136
 0.6229403






R







16
TRIM13
chr13: 5086534: G: A
SLFQSFETWHRGDALSRL
18
17
 0.01839422
 0.6919829



17
CPNE3
chr8: 875541180: G: A
SGQQWYEVEHTERIKNCLN
19
18
 0.08889867
  0.46731711



18
CPQ
chr8: 98155388: G: A
AVVSYVVADMKEMLPRS
17
19
 0.01565236
  0.68498519



19
BDP1
chr5: 70766293: C: T
GQLFPYRARIEKIKNKF
16
20
 0.01549436
  0.71614594



20
ANF784
chr19: 56135881: G: A
ARPEAQSRSSPTLES
15
21
 0.01368863
  0.43412852



21
FOS
chr14: 75748046: T: G
TYPEADSFPSWAAAHRKGS
19
22
 0.06817705
  0.59480153



22
FAMI34A
chr2: 220047223: C: A
FELLDQGELEKLNAELGLEP
20
23
 0.06638302
  0.50878612



23
LTK
chr15: 41796285: G: A
EKLKSWGGSLLGPWLSSGLK
22
24
 0.01248521
  0.57080845






PL







24
SMC2
chr9: 106885498: C: T
YHKQQEELDAFKKTIEESEE
22
25
 0.01245417
  0.52025989






TL







25
UAP1L1
chr9: 139976457: C: T
HGAWLPELPSLPSNGDPPAI
22
26
 0.01167158
  0.48069739






CE







26
ASCC3
chr6: 101296376: A: C
QDDFTALGQMTEKEHG
16
27
 0.01114204
  0.54359088



27
DMAP1
chr1: 44679478: G: A
RDILELGGPEEDAASGTIS
19
28
 0.01011638
 0.5441813



28
NUP98
chr11: 3765801: C: T
GPLGTGAFEAPGFNTTTATL
22
29
 0.00959293
  0.67302464






GF







29
SPOCK1
chr5: 136328211: C: T
FGALHEDANKVIKPTSSNTA
21
30
 0.00848382
 0.4601863






Q







30
EEF1A1
chr6: 74227798: C: T
KSGDAAIVDMVPSKPMCVES
22
31
 0.04120537
0.605315






FS







31
ANF697
chr1: 120166583: C: G
REDDDESAGENPLEEEEEQP
22
32
 0.00771731
  0.41146906






AP







32
MOGS
chr2: 74690506: C: T
SWRVTVEPQDSNTSALPLVS
22
33
 0.00759169
  0.55339174






LF







33
CIC
chr19: 42796535: C: T
PASSQAGTVTLYGPTSSVAL
20
34
 0.00728918
  0.62779866



34
MEPCE
chr7: 100029289: C: T
DGADTSVFSNNVVFVTGNYV
22
35
 0.00681793
  0.70873332






LD







35
MDN1
chr6: 90359807: C: T
WLRRTKPSKHQYQICLAIDD
22
36
 0.00677077
  0.61246852






SS







36
RBM42
chr19: 36120481: T: G
VPGIPTAVPAGPTVPTVPTV
22
37
 0.03277055
 0.4079915






EA







37
MMKI67
chr10: 129901135: G: A
PVKSQSKSNTFLPPLPFKRG
20
38
0.0057615
  0.57937322



38
TEAD3
chr6: 35454323: G: A
GWTTMRRACGARTLSRA
17
39
 0.00532379
  0.68094877



39
FNBP1L
chr1: 94012476: T: C
MGDPGSLQPKSAETMNNIDR
22
40
 0.00510709
  0.44982782






LR







40
EXTL3
chr8: 28574907: C: T
DPRLVIFSGCATRLFE
16
41
 0.00393086
  0.81055314



41
EIF3F
chr11: 8013351: G: A
KHSVKVTNCFSVPHN
15
42
0.0096035
  0.66676886



42
ATP13A1
chr19: 19762504: C: T
EKAHTLILQPPSKKGRQCEW
22
43
 0.00842387
  0.49265976






RS









short
 1
EXOC7
chr17: 74079781: C: T
YIKYKVEQV
 9
44
0.04508725
N/A



 2
LIMA1
chr12: 50571454: G: A
LEDEISKPEV
10
45
0.16698617
N/A



 3
PRKCZ
chr1: 2066754: G: A
YRANSHLF
 8
46
0.01988399
N/A



 4
BST1
chr4: 15716974: C: T
SIKGFFADY
 9
47
0.01757893
N/A



 5
MEGF8
chr19: 42858849: C: T
QRRDRLLTV
 9
48
0.02304197
N/A



 6
CPQ
chr8: 98155388: G: A
YVVADMKEM
 9
49
  0.01129116
N/A
















TABLE 21







Peptide selection for engineering run sample FFFFF




















SEQ





Short/




ID

MHC I
MHC II


Lon
Rank
Gene
Variant
Sequence
NO:
Length
score
score


















long
 1
SLC45A4
chr8: 142264105: G: A
AMETALVTLILLQI
50
17
0.16258827
0.73463601






GKS







 2
PWP2
chr21: 45534552: C: T
KGNIAQMYHAFGKK
51
22
0.14479999
0.68694234






REFNAFVL







 3
PGAP1
chr2: 197744849: G: A
KYLTLRLQDYLSLS
52
20
0.13617044
0.81953448






HLVVYV







 4
MFSD2A
chr1: 40424437: C: A
GRAWDAITDHLVGL
53
20
0.13150237
0.6182176






CISKSP







 5
SLC39A7
chr6: 33171427: C: T
VGSEIAGGAGLGWV
54
22
0.62906294
0.64794417






LPFTAGGF







 6
ZCCHC4
chr4: 25335077: C: T
GFRRVLCVGTLRLH
55
19
0.11540349
0.84718676






ELIKL







 7
THADA
chr2: 43802077: G: A
SAIQVLESSSLSLT
56
18
0.11353184
0.75960565






DSLN







 8
INO80D
chr2: 206921080: G: A
LPLLLFSRAPTVDP
57
16
0.11332213
0.7355256






PR







 9
INO80D
chr2: 206921083: G: A
LPLLLFSRAPTVDP
58
15
0.11291208
0.73622152






P







10
FAM208B
chr10: 5790009: C: T
AKCTGDFSPSLEKL
59
21
0.11199412
0.64350711






VKSGNPL







11
PCM1
chr8: 17813087: C: T
GESNSLTSSVLYPT
60
22
0.55904726
0.80863367






ASLVSQNE







12
COL18A1
chr21: 46910199: G: A
GAKGEVEADGIPGF
61
18
0.52507762
0.57400138






PGLP







13
NUFIP2
chr17: 27613951: G: A
PVDNSSAKIVLKIS
62
21
0.48588871
0.69306097






YASKVKE







14
INTS1
chr7: 1517425: G: A
VVVSSLLLQEEELL
63
22
0.46108
0.62556372






AGGKPGAD







15
NDUFB10
chr16: 2011845: CAG:
ARKCLAKQAEDAAR
64
21
0.46096059
0.49336677





C
EKSCKRG







16
BCAP29
chr7: 107234541: T: G
RNLYISGFSLCFWL
65
22
0.41204663
0.78909355






VLRRLVTL







17
CXorf40B
chrX: 149100859: G: C
KQKYLTVISNRRWL
66
21
0.32483088
0.74806946






LEPIPRK







18
SCARB1
chr12: 125284765: G:
RFSAPLFLSHSHFL
67
21
0.31241171
0.74533032





A
NADPVLA







19
PTPRZ1
chr7: 121694076: C: T
WDHNAQLVVMILDG
68
22
0.30682347
0.65887653






QNMAEDEF







20
TBC1D23
chr3: 100014007: A: G
AIWDGYLQQAGPFF
69
22
0.29311405
0.58318637






IYFLMLII







21
SYNE1
chr6: 152638090: A: C
ASHLEEYNERLELI
70
18
0.28917584
0.56130198






LKWI







22
MBTPS1
chr16: 84115485: G: A
KRELVNSASMKQAL
71
21
0.27998486
0.87108388






IASARRL







23
POLR2B
chr4: 57861015: AT: A
PLDPGGYFIIMDQK
72
16
0.27682839
0.82729797






RF







24
GOLGB1
chr3: 121415783: A: G
LQEALTSRKAIPKK
73
22
0.27224502
0.48823824






AQEKERHL







25
PPP4R2
chr3: 73112870: C: T
LNRMNGVMFSGNSP
74
22
0.23383287
0.77749593






SYTERSNI







26
FARP2
chr2: 242403325: C: T
QRLALWEGPFKAHT
75
19
0.22494294
0.7763395






KGSHQ







27
MRPL38
chr17: 73898232: G: A
REYFGEKTKLKEKI
76
21
0.21683746
0.663767






DIGKPPP







28
FLYWCH1
chr16: 2980828: G: A
PALEEEEAPQALSL
77
22
0.2157846
0.73017404






LSLPPKKR







29
PHF12
chr17: 27244419: G: A
LSNRCQVFDCFQDT
78
20
0.21377716
0.68927787






VSQHVV







30
ITGA7
chr12: 56096837: C: T
DCYRVDIDQEADMQ
79
19
0.16359662
0.5337592






KESKE







31
DDX2
chr20: 47835942: G: A
RRRGGCEKLRAEPQ
80
21
0.15278271
0.78941247






AVLASGS







32
RBL2
chr16: 53499357: A: G
HFYKVIGVFIRAED
81
17
0.14770824
0.83922687






GLC







33
NISCH
chr3: 52526301: C: T
ALTLVFDDVQGYDL
82
20
0.14695032
0.72112204






MGSVTL







34
CDK5RAP3
chr17: 46058540: G: A
MILASPRYVDQVTE
83
22
0.1403607
0.74608702






FLQQKLKQ







35
TANK
chr2: 162061206: C: T
NSTQDNNYGCVSLL
84
22
0.14031428
0.62717317






EDSETRKN







36
SPTBN1
chr2: 54886355: C: T
EEEERKRRPPSLEP
85
22
0.13262047
0.55289858






STKVSEEA







37
VASP
chr19: 46025750: C: T
AGGGPPPAPPLSAA
86
22
0.13190385
0.53188621






QGPGGGGA







38
PRPF
chr6: 4052261: C: T
KLNDADPDDKFYCL
87
22
0.12311099
0.63227453






RLFRHFYH







39
TNPO1
chr5: 72147095: C: T
VQQKLEQLNQYSDF
88
22
0.11718017
0.63499542






NNYLIFVL







40
USP11
chrX: 47104877: C: T
TTVETLEKENSWYC
89
20
0.11154779
0.61009519






PSCKQH







41
RPS24
chr10: 79814617: T: G
VDGDWVLHLPEALS
90
15
0.10728163
0.90107272






A







42
PRRC2B
chr134351481: C: T
RFRRLRQEREFLGL
91
18
0.08403133
0.71237872






WGPE









short
 1
SLC45A4
chr8: 142264105: G: A
METALVTLIL
92
10
0.08417078
N/A



 2
PCM1
chr8: 17813087: C: T
SVLYPTASL
93
 9
0.29510115
N/A



 3
SLC45A4
chr8: 142264105: G: A
ALVTLILLQI
94
10
0.07058771
N/A



 4
COL18A1
chr21: 46910199: G: A
EADGIPGFPGL
95
11
0.28469103
N/A



 5
COL18A1
chr21: 46910199: G: A
GEVEADGIPGF
96
11
0.30583803
N/A



 6
TBC1D9
chr4: 141545323: G: A
RLWTSENKSK
97
10
0.04554415
N/A
















TABLE 22







Peptide Selection for Engineering Run Sample GGGGG





















SEQ




Short/





ID
MCH I
MHC II


Lon
Rank
Gene
Variant
Sequence
Length
NO.
score
score


















long
 1
TSN
chr2: 122519017: G: A
HEHWRFVLQHLVFL
19
 98
0.0464269
0.76456928






AAFVV







 2
MED9
chr17: 17380542: T: C
QSPARAREEENHSF
22
 99
0.02823036
0.84198684






LPLVHNII







 3
RBM28
chr7: 127975934: G: A
EDEEEENIELKVTK
17
100
0.12394809
0.50573551






PVQ







 4
PRKDC
chr8: 48690396: G: A
PELMPFRFLTCQFI
21
101
0.07648852
0.76358768






NLMLPMKE







 5
ZNF12
chr7: 6737368: C: G
EEWQQLDPEQNITY
22
102
0.01395768
0.54926563






RDVMLENY







 6
MRPL48
chr11: 73575383: G: A
DFKGRFKARPKLEE
20
103
0.01373842
0.6212347






LLAKLK







 7
INTS1
chr7: 1526709: G: A
ASSQSMPWLVDLVQ
22
104
0.05733778
0.62354692






SSEGSLDV







 8
DNAJC10
chr2: 183622543: A: G
RFFPPKSNKACHYH
21
105
0.05379868
0.63486407






SYNGWNR







 9
MEN1
chr11: 64573759: G: A
MYLAGYHCRNCNVR
21
106
0.00744509
0.8061702






EALQAWA







10
SNX25
chr4: 186185720: C: T
LVFCLLPSKDVQFL
15
107
0.00702811
0.83040282






S







11
LCMT1
chr16: 25180500: C: T
RRRQCDLVGVETCK
19
108
0.00388609
0.65396226






SLESQ







12
INF2
chr14: 105179557: G:
FLRALKENKDQKEQ
21
109
0.0161754
0.43941237





A
AAKAERR









short
 1
RSN
chr2: 122519017: G: A
FVLQHLVFL
 9
110
0.03300495
N/A



 2
MED9
chr17: 17380542: T: C
REEENHSFL
 9
111
0.01610038
N/A



 3
MED9
chr17: 17380542: T: C
REEENHSFLPL
11
112
0.01619894
N/A



 4
TSN
chr2: 122519017: G: A
VLQHLVFLA
 9
113
0.01469447
N/A



 5
MED9
chr17: 17380542: T: C
EENHSFLPL
 9
114
0.00951051
N/A



 6
INTS1
chr7: 1526709: G: A
SMPWLVDLV
 9
115
0.04252224
N/A



 7
MED9
chr17: 17380542: T: C
RAREEENHSF
10
116
0.0088703
N/A



 8
MRPL48
chr11: 73575383: G: A
KARPKLEEL
 9
117
0.00901679
N/A



 9
TSN
chr2: 122519017: G: A
FVLQHLVFLA
10
118
0.00723975
N/A



10
PRKDC
chr8: 48690396: G: A
RLTCQFINL
 9
119
0.03565368
N/A



11
ZNF12
chr7: 6737368: C: G
QLDPEQNITY
10
120
0.0119429
N/A



12
RBM28
chr7: 127975934: G: A
NIELKVTKPVQ
11
121
0.05185511
N/A



13
MED9
chr17: 17380542: T: C
REEENHSF
 8
122
0.00539441
N/A



14
MED9
chr17: 17380542: T: C
EEENHSFLPL
10
123
0.00433009
N/A



15
RBM28
chr7: 127975934: G: A
NIELKVTKPV
10
124
0.03534479
N/A



16
TSN
chr2: 122519017: G: A
FVLQHLVF
 8
125
0.0063978
N/A



17
RBM28
chr7: 127975934: G: A
DEEEENIELKV
11
126
0.04180784
N/A



18
RBM28
chr7: 127975934: G: A
EEEENIELKV
10
127
0.03181995
N/A



19
MED9
chr17: 17380542: T: C
RAREEENHSFL
11
128
0.00303641
N/A



20
TSN
chr2: 122519017: G: A
VLQHLVFLAA
10
129
0.0029588
N/A



21
RBM28
chr7: 127975934: G: A
EEEENIELKVT
11
130
0.02003145
N/A



22
INTS1
chr7: 1526709: G: A
QSMPWLVDLV
10
131
0.01398635
N/A



23
PRKDC
chr8: 48690396: G: A
LTCQFINLM
 9
132
0.01486982
N/A



24
RBM28
chr7: 127975934: G: A
EEENIELKVTK
11
133
0.01899035
N/A



25
TSN
chr2: 122519017: G: A
VLQHLVFL
 8
134
0.00307205
N/A



26
SNX25
chr4: 186185720: C: T
LLPSKDVQFL
10
135
0.00218817
N/A



27
RBM28
chr7: 127975934: G: A
EDEEEENIELK
11
136
0.00853576
N/A



28
ZNF12
chr7: 6737368: C: G
QLDPEQNITYR
11
137
0.00213218
N/A



29
INF2
chr14: 105179557: G:
ALEKNKDQK
 9
138
0.00667427
N/A





A








30
MRPL48
chr11: 73575383: G: A
KLEELLAK
 8
139
0.0020238
N/A



31
MED9
chr17: 17380542: T: C
AREEENHSFLP
11
140
0.00150676
N/A



32
DNAJC10
chr2: 183622543: A: G
KSNKACHY
 8
141
0.01944355
N/A



33
RBM28
chr7: 127975934: G: A
PSKDVQFL
 8
142
0.00137019
N/A



34
PADRE
N/A
EEEENIELK
 9
143
0.00556938
N/A




PADRE
N/A
AKFVAAWTLKAAA
13
144
N/A
N/A






AKFVAAWTLKAAA
13
145
N/A
N/A









1.4. Peptide Manufacturing

Vaccine peptide manufacturing and pool formulation occured at a peptide manufacturer performing peptide synthesis, quality control, dissolving and mixing of peptides and peptide pools.


Peptides were prepared by solid phase peptide synthesis, purified using RP-HPLC columns, and analyzed for quality (identity, purity, peptide content, Acetate/TFA content, residual organic solvents).


Results of the peptide synthesis are shown in Tables 23 to 25.









TABLE 23







Peptide Synthesis Engineering Run Batch for Sample DDDDD_2020

















Peptide
Peptide

SEQ

Peptide
Final
Passed



No.
ID
Lot
Peptide Sequence
ID NO:
Length
Classss
Lyo
QC
Solubility



















 1
DDDDD.1
DDDDD.1
PASSQAGTVTLYGPTSSVALGF
146
25
Class
N/A
N/A
N/A





TSL


I/II








 2
DDDDD.2
DDDDD.2
EDDDESAGENPLEEEEEQPAP
147
21
Class
N/A
N/A
N/A








I/II








 3
DDDDD.3
DDDDD.2
MSKPKWPLEDEISKPEVP
148
18
Class
Y
Y
Clear








I/II








 4
DDDDD.4
DDDDD.4
NQPKIGGPLGTGAFEAPGFNTT
149
28
Class
Y
Y
Clear





TATLGF


I/II








 5
DDDDD.5
DDDDD.5
FGALHEDANKVIKPTSSNTA
150
20
Class
Y
Y
Clear








I/II








 6
DDDDD.6
DDDDD.6
KNPQMGDPGSLQPKSAETMNN
151
28
Class
Y
Y
Clear





IDRLRME


I/II








 7
DDDDD.7
DDDDD.7
LQMLCPQEAPQILSRLEAVR
152
20
Class
Y
Y
Clear








I/II








 8
DDDDD.8
DDDDD.8
YVMVVMSDSSIPSAATLINIRN
153
23
Class
N/A
N/A
N/A





A


I/II








 9
DDDDD.9
DDDDD.9
SSCMGGMNQRPILTIITLED
154
20
Class
Y
Y
Cloudy








I/II








10
DDDDD.10
DDDDD.10
YRAPEIILGLLFCEAI
155
16
Class
N/A
N/A
N/A








I/II








11
DDDDD.11
DDDDD.11
DPRLVIFSGCATRLFEA
156
17
Class
Y
Y
Cloudy








I/II








12
DDDDD.12
DDDDD.12
ARPEAQSRSSPTLES
157
15
Class
Y
Y
Clear








I/II








13
DDDDD.13
DDDDD.13
DGADTSVFSNNVVFVT
158
16
Class
N/A
N/A
N/A








I/II








14
DDDDD.14
DDDDD.14
GQRRDRLLTVQALSGL
159
16
Class
Y
Y
Clear








I/II








15
DDDDD.15
DDDDD.15
WLPELPSLPSNGDPPAICE
160
19
Class
Y
Y
Clear








I/II








16
DDDDD.16
DDDDD.16
LLGTVDKHSVKVTNCFSVPHN
161
21
Class
Y
Y
Clear








I/II








17
DDDDD.17
DDDDD.17
KDKIWLRRTKPSKHQYQICLAI
162
28
Class
Y
Y
Clear





DDSSSM


I/II








18
DDDDD.18
DDDDD.18
QELSPEKLKSWGGSLLGPWLSS
163
28
Class
Y
Y
Clear





GLKPLK


I/II








19
DDDDD.19
DDDDD.19
ESADLPPKGFQASYGKDE
164
18
Class
N/A
N/A
N/A








I/II








20
DDDDD.20
DDDDD.20
TYPEADSFPSWAAAHRKGSS
165
20
Class
Y
Y
Clear








I/II








21
DDDDD.21
DDDDD.21
RDAFESLFQSFETWHRGDALSR
166
24
Class
Y
Y
Semi-





LD


I/II


cloudy





22
DDDDD.22
DDDDD.22
APPTRLAPPQRPPPPS
167
16
Class
Y
Y
Clear








I/II








23
DDDDD.23
DDDDD.23
MWSSINCIICACVKGR
168
16
Class
N/A
N/A
N/A








I/II








24
DDDDD.24
DDDDD.24
RRWRKLYRANSHLFQAKRFN
169
20
Class
Y
Y
Clear








I/II








25
DDDDD.25
DDDDD.25
AVVSYVVADMKEMLPRS
170
17
Class
Y
Y
Clear








I/II








26
DDDDD.26
DDDDD.26
LNGSEPTGAYSIKGFFADYEIP
171
23
Class
Y
Y
Clear





N


I/II








27
DDDDD.27
DDDDD.27
GQLFPYRARIEIKNKF
172
16
Class
Y
Y
Clear








I/II








28
DDDDD.28
DDDDD.28
QFDQLYRTKPGMTMSC
173
16
Class
N/A
N/A
N/A








I/II








29
DDDDD.29
DDDDD.29
PRTESSDVADQLWAQ
174
15
Class
Y
Y
Semi-








I/II


cloudy





30
DDDDD.30
DDDDD.30
RDILELGGPEEDAASGTISKKD
175
22
Class
Y
Y
Clear








I/II








31
DDDDD.31
DDDDD.31
SQAGTVTL
176
 8
Class
Y
Y
Clear








I








32
DDDDD.32
DDDDD.32
QSRSSPTL
177
 8
Class
N/A
N/A
N/A








I








33
DDDDD.33
DDDDD.33
KVTNCFSV
178
 8
Class
Y
Y
Clear








I








34
DDDDD.34
DDDDD.34
TWHRGDAL
179
 8
Class
Y
Y
Clear








I








35
DDDDD.35
DDDDD.35
SSQAGTVTL
180
 9
Class
Y
Y
Semi-








I


cloudy





36
DDDDD.36
DDDDD.36
AQSRSSPTL
181
 9
Class
Y
Y
Clear








I








37
DDDDD.37
DDDDD.37
RRACGARTL
182
 9
Class
Y
Y
Clear








I








38
DDDDD.38
DDDDD.38
ASSQAGTVTL
183
10
Class
Y
Y
Clear








I








39
DDDDD.39
DDDDD.39
RAPEIILGLL
184
10
Class
Y
Y
Semi-








I


cloudy





40
DDDDD.40
DDDDD.40
EAQSRSSPTL
185
10
Class
Y
Y
Clear








I








41
DDDDD.41
DDDDD.41
NTSALPLVSL
186
10
Class
Y
Y
Clear








I








42
DDDDD.42
DDDDD.42
DSSIPSAATL
187
10
Class
Y
Y
Clear








I








43
DDDDD.43
DDDDD.43
SAGENPLEEEE
188
11
Class
Y
Y
Clear








I








44
DDDDD.44
DDDDD.44
NSPSTPTEQRI
189
11
Class
N/A
N/A
N/A








I








45
DDDDD.45
DDDDD.45
FEAPGFNTTTA
190
11
Class
Y
Y
Clear








I








46
DDDDD.46
DDDDD.46
AKFVAAWTLKAAA
191
13
PADRE
Y
Y
Clear





47
DDDDD.47
DDDDD.
AKFVAAWTLKAAA
192
13
PADRE
Y
Y
Clear




46-1












48
DDDDD.48
DDDDD.
AKFVAAWTLKAAA
193
13
PADRE
Y
Y
Clear




46-2























TABLE 24







Peptide synthesis engineering run batch for sample FFFFF

















Peptide
Peptide

SEQ

Peptide
Final
Passed



No.
ID
Lot
Peptide Sequence
ID NO:
Length
Class
Lyo
QC
Solubility



















 1
FFFFF.1
FFFFF.1
LFMLTFSTSPGLESPVESFIAFLLI
194
25
Class
N/A
N/A
N/A








I/II








 2
FFFFF.2
FFFFF.2
PVVNGESNSLTSSVLYPTASLVSQN
195
25
Class
N/A
N/A
N/A








I/II








 3
FFFFF.3
FFFFF.3
SARKCLAKQAEDAAREKSCK
196
20
Class
N/A
N/A
N/A








I/II








 4
FFFFF.4
FFFFF.4
TNSAIQVLESSSLSLTDSLNGNS
197
23
Class
N/A
N/A
N/A








I/II








 5
FFFFF.5
FFFFF.5
PTLPQPASHFSPPPPPPPLPP
198
21
Class
Y
Y
Clear








I/II








 6
FFFFF.6
FFFFF.6
FINPIFEFSQAMRRLGLDDA
199
20
Class
Y
N
N/A








I/II








 7
FFFFF.7
FFFFF.7
STVQKRELVNSASMKQALIASARRLP
200
26
Class
Y
N/A
N/A








I/II








 8
FFFFF.8
FFFFF.8
EDTGQDMLALFLRTNRQAAK
201
20
Class
Y
Y
Clear








I/II








 9
FFFFF.9
FFFFF.9
YGRTVVPFLVPGTSQLGQ
202
18
Class
Y
Y
Clear








I/II








10
FFFFF.10
FFFFF.10
YIHTSVSQDFSQSVPGTTSSPL
203
22
Class
Y
Y
Clear








I/II








11
FFFFF.11
FFFFF.11
VVVSSLLLQEEELLAGGKPGADG
204
23
Class
Y
N
N/A








I/II








12
FFFFF.12
FFFFF.12
LSNQQPGLMVSFSLRLFPLFV
205
21
Class
N/A
N/A
N/A








I/II








13
FFFFF.13
FFFFF.13
PVDNSSAKIVLKISYASKVKE
206
21
Class
N/A
N/A
N/A








I/II








14
FFFFF.14
FFFFF.14
QIMLRSGVDLSVTDKREWRP
207
20
Class
Y
N
N/A








I/II








15
FFFFF.15
FFFFF.15
RNQHQRLLKNMGAHLVVLDLLQIPYE
208
26
Class
N/A
N/A
N/A








I/II








16
FFFFF.16
FFFFF.16
QPASAAKCTGDFSPSLEKLVKSGNPL
209
30
Class
N/A
N/A
N/A





QPVS


I/II








17
FFFFF.17
FFFFF.17
TPSAPEGYDLKIGLFLAPRRGSLPD
210
25
Class
Y
Y
Clear








I/II








18
FFFFF.18
FFFFF.18
MILASPRYVDQVTEFLQQKLK
211
21
Class
Y
Y
Clear








I/II








19
FFFFF.19
FFFFF.19
VGRAWDAITDHLVGLCISKSP
212
21
Class
Y
Y
Clear








I/II








20
FFFFF.20
FFFFF.20
FLSPGQLLQEPRTSLLIINNT
213
21
Class
Y
Y
Cloudy








I/II








21
FFFFF.21
FFFFF.21
RSTTKSPGPSRHSKSPASTSSVN
214
23
Class
Y
Y
Clear








I/II








22
FFFFF.22
FFFFF.22
KLESTVGSPKKPLSDLGKLS
215
20
Class
Y
Y
Clear








I/II








23
FFFFF.23
FFFFF.23
GRPRMMGTGLSPYPEHLTSPLSPAQ
216
25
Class
Y
N
N/A








I/II








24
FFFFF.24
FFFFF.24
PLNPPASTAFSQEPHSGSPA
217
20
Class
Y
Y
Clear








I/II








25
FFFFF.25
FFFFF.25
AAFVTPDQKYSMDNTLHTPTPFKNAL
218
27
Class
Y
N
N/A





E


I/II








26
FFFFF.26
FFFFF.26
RNLYISGFSLCFWLVLRRLVT
219
21
Class
Y
Y
Cloudy








I/II








27
FFFFF.27
FFFFF.27
NAQLVVMILDGQNMAEDEF
220
19
Class
Y
N/A
N/A








I/II








28
FFFFF.28
FFFFF.28
RVLCVGTLRLHELIKLTA
221
18
Class
Y
N/A
N/A








I/II








29
FFFFF.29
FFFFF.29
HKDAVTCVNFSSSGHLLASGSR
222
22
Class
Y
Y
Clear








I/II








30
FFFFF.30
FFFFF.30
LGNLAQFWECCLSSSGDADGESF
223
23
Class
N/A
N/A
N/A








I/II








31
FFFFF.31
FFFFF.31
HIAGTSGFSLSFHSTVIN
224
18
Class
N/A
N/A
N/A








I/II








32
FFFFF.32
FFFFF.32
LPTIKYLTLRLQDYLSLSHLVVYVPS
225
26
Class
N/A
N/A
N/A








I/II








33
FFFFF.33
FFFFF.33
RLVHSGSGCRSPFLGSDLTFATRTGS
226
28
Class
N/A
N/A
N/A





RQ


I/II








34
FFFFF.34
FFFFF.34
PPKSPGPHSEKEDEAEPSTVPG
227
22
Class
Y
N
N/A








I/II








35
FFFFF.35
FFFFF.35
PFLHTVSKTRLFEYLRLTSL
228
20
Class
Y
Y
Semi-








I/II


Cloudy





36
FFFFF.36
FFFFF.36
TGQDMLAL
229
 8
Class I
Y
Y
Clear





37
FFFFF.37
FFFFF.37
YKTDLHSL
230
 8
Class I
Y
Y
Clear





38
FFFFF.38
FFFFF.38
LAQFWECCL
231
 9
Class I
Y
Y
Cloudy





39
FFFFF.39
FFFFF.39
SLLLQEEEL
232
 9
Class I
Y
Y
Cloudy





40
FFFFF.40
FFFFF.40
CTGDFSPSL
233
 9
Class I
Y
N
N/A





41
FFFFF.41
FFFFF.41
AQFWECCL
234
 8
Class I
Y
Y
Cloudy





42
FFFFF.42
FFFFF.42
LLLQEEEL
235
 8
Class I
Y
Y
Clear





43
FFFFF.43
FFFFF.43
TGDFSPSL
236
 8
Class I
Y
Y
Semi-











Cloudy





44
FFFFF.44
FFFFF.44
QVLESSSL
237
 8
Class I
Y
N
N/A





45
FFFFF.45
FFFFF.45
NSLTSSVL
238
 8
Class I
Y
Y
Clear





46
FFFFF.46
FFFFF.46
NSSAKIVL
239
 8
Class I
Y
Y
Semi-











Cloudy





47
FFFFF.47
FFFFF.47
NSASMKQAL
240
 9
Class I
Y
Y
Clear





48
FFFFF.48
FFFFF.48
AIQVLESSSL
241
10
Class I
Y
Y
Semi-











Cloudy





49
FFFFF.49
FFFFF.49
LGIGGLQDL
242
 9
Class I
Y
Y
Clear





50
FFFFF.50
FFFFF.50
LSPYPEHL
243
 8
Class I
Y
Y
Clear





51
FFFFF.51
FFFFF.51
AKFVAAWTLKAAA
244
13
PADRE
Y
Y
Clear





52
FFFFF.51
FFFFF.51-1
AKFVAAWTLKAAA
245
13
PADRE
Y
N/A
N/A
















TABLE 25







Peptide Synthesis Engineering Run Batch for Sample GGGGG

















Peptide
Peptide

SEQ

Peptide
Final
Passed
Stability


No.
ID
Lot
Peptide Sequence
ID NO:
Length
Class
Lyo
QC




















 1
GGGGG.1
GGGGG.1
SPRVAPGSAPPWPALRSLLH
246
20
Class
Y
Y
Clear








I/II








 2
GGGGG.2
GGGGG.2
VLGTSAPGSSRLAAVDLGG
247
19
Class
Y
Y
Clear








I/II








 3
GGGGG.3
GGGGG.3
ARPPGGSGPLRVLIPDLQL
248
19
Class
Y
Y
Clear








I/II








 4
GGGGG.4
GGGGG.4
PSAPQQEGVASKEKEEV
249
17
Class
Y
Y
Clear








I/II








 5
GGGGG.5
GGGGG.5
TSQAYNALTLVVTSCKNFKVRI
250
22
Class
N/A
N/A
N/A








I/II








 6
GGGGG.6
GGGGG.6
GENSVSSSPSASSTAALNTAAA
251
22
Class
N/A
N/A
N/A








I/II








 7
GGGGG.7
GGGGG.7
FETTTGFDPHSGTPLSDHEAL
252
21
Class
Y
Y
Clear








I/II








 8
GGGGG.8
GGGGG.8
FQSLCQAPPLLKDKVLTALE
253
20
Class
Y
Y
Clear








I/II








 9
GGGGG.9
GGGGG.9
VVPGNVTLSVVGSTSVPLSS
254
20
Class
N/A
N/A
N/A








I/II








10
GGGGG.10
GGGGG.10
RPGEDPSLHGIVKEQL
255
16
Class
Y
Y
Clear








I/II








11
GGGGG.11
GGGGG.11
SPAESCDLLGAIQTCIRKSLG
256
21
Class
Y
Y
Clear








I/II








12
GGGGG.12
GGGGG.12
MKMASFLAFLLLNFHVCLLLLQ
257
25
Class
N/A
N/A
N/A





LLM


I/II








13
GGGGG.13
GGGGG.13
SENQQPGAPNTPTHPAPPGLH
258
21
Class
N/A
N/A
N/A








I/II








14
GGGGG.14
GGGGG.14
HLINYQDDAELATHALPELTKL
259
29
Class
Y
Y
Clear





LNDEDPV


I/II








15
GGGGG.15
GGGGG.15
LIVENVHFQAHKALLAA
260
17
Class
Y
Y
Semi








I/II


Cloudy





16
GGGGG.16
GGGGG.16
STAPAEATLPKPGEAEAP
261
18
Class
Y
Y
Clear








I/II








17
GGGGG.17
GGGGG.17
PYSGLGGVGDPYAPLMVLMCR
262
26
Class
Y
Y
Semi





VCLED


I/II


Cloudy





18
GGGGG.18
GGGGG.18
DFYLRGAVALSVRPIS
263
16
Class
N/A
N/A
N/A








I/II








19
GGGGG.19
GGGGG.19
TDVDPQSAVMQEEIF
264
15
Class
N/A
N/A
N/A








I/II








20
GGGGG.20
GGGGG.20
NSLQNQALQTLQERLHEADAT
265
21
Class
Y
Y
Clear








I/II








21
GGGGG.21
GGGGG.21
GAEDSIDSPSACPLSTGCPAL
266
21
Class
Y
Y
Clear








I/II








22
GGGGG.22
GGGGG.22
RFIGPLPREGSVGSTSDYVSQS
267
22
Class
Y
Y
Clear








I/II








23
GGGGG.23
GGGGG.23
IQSIYGGLPKVPAKPKEPTIPH
268
22
Class
Y
Y
Clear








I/II








24
GGGGG.24
GGGGG.24
FWGILGFPALYTHLPAFLEWTL
269
26
Class
Y
N
N/A





CLLS


I/II








25
GGGGG.25
GGGGG.25
VDLKFPASVPTGAQDLISKL
270
20
Class
Y
Y
Clear








I/II








26
GGGGG.26
GGGGG.26
LAPGQPFLSSQGSLCI
271
16
Class
N/A
N/A
N/A








I/II








27
GGGGG.27
GGGGG.27
RVGDLSPKQKEALAKPEA
272
18
Class
Y
Y
Clear








I/II








28
GGGGG.28
GGGGG.28
LAVRWFFAHSSDSQEALMV
273
19
Class
Y
N
N/A








I/II








29
GGGGG.29
GGGGG.29
YSGIQESSSASPLSIKKCPI
274
20
Class
Y
Y
Clear








I/II








30
GGGGG.30
GGGGG.30
LIKPPAHTSAILTVLR
275
16
Class
Y
Y
Clear








I/II








31
GGGGG.31
GGGGG.31
MSYELKCAQELSQKQDG
276
17
Class
Y
Y
Clear








I/II








32
GGGGG.32
GGGGG.32
QVHQCSVLLVATGLSVP
277
17
Class
N/A
N/A
N/A








I/II








33
GGGGG.33
GGGGG.33
RSLTLEPDPIVVPGNVTLSWG
278
22
Class
Y
Y
Clear








I/II








34
GGGGG.34
GGGGG.34
LDRQHVQHQLLVILKELRK
279
19
Class
Y
Y
Clear








I/II








35
GGGGG.35
GGGGG.35
TVDMLQCLRFPGLALPHTRAPS
280
25
Class
Y
Y
Clear





PLG


I/II








36
GGGGG.36
GGGGG.36
SAPGSSRLAAV
281
11
Class
Y
Y
Clear








I








37
GGGGG.37
GGGGG.37
VALSVRPI
282
 8
Class
Y
Y
Clear








I








38
GGGGG.38
GGGGG.38
SAVMQEEI
283
 8
Class
Y
Y
Cloudy








I








39
GGGGG.39
GGGGG.39
AAIQEKKEI
284
 9
Class
Y
Y
Clear








I








40
GGGGG.40
GGGGG.40
VSPDIFMQSHL
285
11
Class
Y
Y
Clear








I








41
GGGGG.41
GGGGG.41
QAYNALTL
286
 8
Class
Y
Y
Clear








I








42
GGGGG.42
GGGGG.42
RVLIPDLQL
287
 9
Class
Y
Y
Clear








I








43
GGGGG.43
GGGGG.43
APGSSRLA
288
 8
Class
Y
Y
Clear








I








44
GGGGG.44
GGGGG.44
STAPAEATL
289
 9
Class
Y
Y
Clear








I








45
GGGGG.45
GGGGG.45
GALPVASPASL
290
11
Class
Y
Y
Clear








I








46
GGGGG.46
GGGGG.46
SAPGSSRLAA
291
10
Class
Y
Y
Clear








I








47
GGGGG.47
GGGGG.47
TSAPGSSRLAA
292
11
Class
Y
Y
Clear








I








48
GGGGG.48
GGGGG.48
SLCQAPPL
293
 8
Class
Y
Y
Clear








I








49
GGGGG.49
GGGGG.49
MSYELKCAQEL
294
11
Class
Y
Y
Clear








I








50
GGGGG.50
GGGGG.50
LAPGQPFL
295
 8
Class
y
Y
Clear








I








51
GGGGG.51
GGGGG.51
AKFVAAWTLKAAA
296
13
PADRE
Y
Y
Clear





52
GGGGG.52
GGGGG.51-1
AKFVAAWTLKAAA
297
13
PADRE
N/A
N/A
N/A









Peptides that were successfully manufactured as determined by passing quality control criteria (appearance, identity, peptide content, peptide purity, acetate content, TFA content, residual organic solvents), were then ingested into a pool optimization algorithm.


Briefly, this algorithm distributed the selected peptide to 4 pools, optimizing distribution so that each pool contains peptides with both high MHC class I and class II scores. After initial exclusion of sequences which contain more than two cysteines to avoid multimerization during pool formulation, an optimization algorithm identified the best combination of approximately 14 long vaccine peptides and 6 short peptides to be combined into individual vaccine peptide pools. If the initial peptide prediction or manufacturing yielded less than 14 long vaccine peptides, the long—to—short peptide ratio may be changed to accommodate the target of four pools with five peptides each. Similarly, if a vaccine peptide pool was composed of less than three (3) long peptides, which extrapolates to a low chance of CD4 engagement, one of the pool peptides was chosen to be PADRE.


Several proposed pool compositions were then communicated to the peptide manufacturer, and vaccine peptide pool formulation is performed:


Groups of peptides are selected and pooled. Up to four (4) pools with not more than (NMT) five (5) peptides in each pool were prepared. Each peptide was dissolved in 5.538% (v/v) DMSO followed by 0.9% NaCl solution, at a concentration of each peptide at 0.4158 mg/mL. Peptides which were visibly dissolved were accepted for that group's pool. Upon successful pooling of all peptides within a group, the pool was filtered through a 0.2 μm Nylon filter. Peptides were shipped in sterile tubes and labeled with Pool name, lot #, components, manufacturing data, quantity/concentration and storage conditions.


1.5. Formulation of Peptide Vaccine

To formulate a personalized vaccine, peptide pools were admixed with Poly ICLC as adjuvant using the following procedure:


Remove one peptide vial each per pool from the freezer. The following steps are performed for each of the peptide pools.


Thaw the peptide pools for 20 to 30 min at room temperature. The peptide solution contains DMSO and may require slight warming (hand-warm) and/or agitation for complete thawing. The substance is expected to be a clear, colorless solution. If precipitates form, the solution may be vortexed to resolve precipitates.


While thawing, prepare and label syringes for steps 4-9 (see Table 26). (A) Two (2) mixing syringes per pool, labeled “M1-2 [A-D]” (denoting pools): (i) Syringe 1: 10 mL (or appropriately sized) and (ii) Syringe 2: 1 mL (or appropriately sized); and (B) One (1) administration syringe per pool, labeled “administration syringe [A-D]” (denoting pools).









TABLE 26







List of syringes used for preparation and administration.










Suggested




Syringe



Syringe Labela
Volumeb
Syringe Use





M1_[pool name]
10 mL
Aspiration of peptide pool, sterile filtration


M2_[pool name]
 1 mL
Aspiration Poly ICLC, transfer into M1


A1_[pool name]
 1 mL
Receiving Syringe for final drug product,




Administration






aAll syringes are to be prepared once each per peptide pool




bUse suggested volume syringe or appropriate size







Additional Material Required (Per Peptide Pool)

1 (one) guarded female to female Luer adaptor


1 (one) 3″ aspiration needle to transfer peptide pool into mixing syringe M1


1 (one) sterile low-protein binding syringe filter, non-pyrogenic. Pore size 0.22 μm. (e.g. Pall DMSO-Safe Aerodisc Syringe Filter, #4433, or Millex-GV 0.22 μm PVDF, 33 mm, gamma sterilized. Millipore, #SLGVM33RS)


3 (three) Sterile Hypodermic needle appropriate gauge


1 (one) Spiros Closed System Drug-Transfer Device (CSTD) for IM injection [ICU Medical, #SH2000SC-10]


Sterile filter the content of each vial: Using the 10mL syringe with Luer Lock (mixing syringe 1, M1), connected to the 0.22 μm sterile filter, slide the 3″ aspiration needle on the assembly, and remove 2mL peptide pool from the vial.


Prepare Poly ICLC solution. Using a 1mL syringe (mixing syringe 2, M2) and appropriately sized needle, aspirate 0.760 mL of Poly ICLC under sterile conditions. Poly ICLC is a white opalescent solution. Downstream steps may lead to an opalescent product, which is acceptable.


Remove the needles from mixing syringe 1 (M1) and mixing syringe 2 (M2), as well as sterile filter from mixing syringe 1 (M1).


Connect Poly ICLC mixing syringe 2 (M2) with the peptide pool mixing syringe 1 (M1) via a female-to-female Luer Lock guarded connector


Transfer Poly ICLC mixing syringe 2 (M2) into mixing syringe 1 (M1) and mix diligently between both syringes. Bubbles may form in the product. Tapping the syringe may help to collect bubbles. The resultant mixture is the personalized vaccine.

Claims
  • 1. A method, comprising: a) obtaining sequence data from the tumor, wherein the sequence data is used to obtain data representing a polypeptide sequence of one or more tumor-specific neoantigens;b) obtaining data representing MHC molecule polypeptide sequence data from the subject;c) inputting the data representing the polypeptide sequence(s) and the data representing the MHC molecule(s) polypeptide sequence(s) into a machine-learning platform to generate a numerical probability score that the one or more tumor-specific neoantigens will elicit an immune response in the subject;d) quantifying RNA expression of the one or more tumor-specific neoantigens in a tumor to identify one or more tumor-specific neoantigens that express an amount of the one or more tumor-specific neoantigens sufficient to elicit an immune response in the subject;e) calculating a tumor-specific neoantigen score based on steps c) and d) for the one or more tumor-specific neoantigens; andf) selecting one or more tumor-specific neoantigens based on the tumor-specific score for formulation of a subject-specific immunogenic composition.
  • 2. The method of claim 1, further comprising: g) forming a subject-specific immunogenic composition comprising the one or more selected tumor-specific neoantigens; andh) administering the immunogenic composition to the subject.
  • 3. The method of claim 1, further comprising, before step e), sequencing tumor clones of the tumor to identify one or more tumor-specific neoantigens that represent a sufficient fraction of the tumor.
  • 4. The method of claim 3, wherein the tumor-specific neoantigen score is calculated based on steps c), d), and the sequencing the tumor clones.
  • 5. The method of claim 1, wherein the polypeptide sequence of the one or more tumor-specific neoantigens is from short polypeptides.
  • 6. (canceled)
  • 7. The method of claim 1, wherein the polypeptide sequence of the one or more tumor-specific neoantigens is from long polypeptides.
  • 8-13. (canceled)
  • 14. The method of claim 1, wherein a tumor-specific neoantigen with a higher numerical probability score relative to a lower numerical probability score indicates that the tumor-specific neoantigen will elicit a greater immune response in the subject.
  • 15. (canceled)
  • 16. The method of claim 1, wherein the quantifying the RNA expression comprises measuring mRNA expression.
  • 17. The method of claim 3, wherein the one or more tumor-specific neoantigens represent at least about 1% of the tumor.
  • 18. The method of claim 3, wherein the one or more tumor-specific neoantigens represent at least about 5% of the tumor.
  • 19. The method of claim 1, wherein the sequence data is nucleotide sequence data.
  • 20. The method of claim 1, wherein the sequence data is polypeptide sequence data.
  • 21. (canceled)
  • 22. The method of claim 1, wherein at least about 10 tumor-specific neoantigens are selected to formulate the subject-specific immunogenic composition.
  • 23. The method of claim 22, wherein at least about 20 tumor-specific neoantigens are selected to formulate the subject-specific immunogenic composition.
  • 24-25. (canceled)
  • 26. The method of claim 1, wherein the immunogenic composition comprises one or more tumor-specific neoantigens encoded by short polypeptides.
  • 27. The method of claim 1, wherein the immunogenic composition comprises one or more tumor-specific neoantigens encoded by long polypeptides.
  • 28. (canceled)
  • 29. The method of claim 1, further comprising identifying one or more tumor-specific neoantigens that induce an autoimmune response to normal tissue.
  • 30. The method of claim 29, wherein, in step e), the one or more tumor-specific neoantigens that induce an autoimmune response to normal tissue are not selected for the immunogenic composition.
  • 31. The method of claim 29, wherein the one or more tumor-specific neoantigens that induce an autoimmune response to normal tissue has a lower tumor-specific neoantigen score relative to a tumor-specific neoantigen that does not induce an autoimmune response.
  • 32. The method of claim 1, wherein the tumor is from melanoma, breast cancer, ovarian cancer, prostate cancer, kidney cancer, gastric cancer, colon cancer, testicular cancer, head and neck cancer, pancreatic cancer, brain cancer, B-cell lymphoma, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, T-cell lymphocytic leukemia, bladder cancer, or lung cancer.
  • 33. The method of claim 32, wherein the tumor is from a cancer selected from the group consisting of a melanoma, breast cancer, lung cancer, and bladder cancer.
  • 34-41. (canceled)
  • 42. The method of claim 1, further comprising formulating an immunogenic composition comprising the selected one or more tumor-specific neoantigens.
  • 43. An immunogenic composition comprising one or more tumor-specific neoantigens selected by performing the method of claim 1.
  • 44. The immunogenic composition of claim 43, wherein the immunogenic composition comprises a nucleotide sequence, a polypeptide sequence, RNA, DNA, a cell, a plasmid, a vector, a dendritic cell, or a synthetic long peptide.
  • 45. The immunogenic composition of claim 43, further comprising an adjuvant.
  • 46. The method of claim 1, wherein the sequence data is whole exome sequence data, RNA sequence data, whole genome sequence data or combinations thereof.
  • 47. (canceled)
  • 48. A method for treating cancer in a subject in need thereof, comprising: a) obtaining sequence data from the tumor, wherein the sequence data is used to obtain data representing a polypeptide sequence of one or more tumor-specific neoantigens;b) obtaining data representing MHC molecule polypeptide sequence data from the subject;c) inputting the data representing the polypeptide sequence(s) and the data representing the MHC molecule(s) polypeptide sequence(s) into a machine-learning platform to generate a numerical probability score that the one or more tumor-specific neoantigens will elicit an immune response in the subject;d) quantifying RNA expression of the one or more tumor-specific neoantigens in a tumor to identify one or more tumor-specific neoantigens that express an amount of the one or more tumor-specific neoantigens sufficient to elicit an immune response in the subject;e) calculating a tumor-specific neoantigen score based on steps c) and d) for the one or more tumor-specific neoantigens; andf) selecting one or more tumor-specific neoantigens based on the tumor-specific score for formulation of a subject-specific immunogenic composition.g) forming a subject-specific immunogenic composition comprising the one or more selected tumor-specific neoantigens; andh) administering the immunogenic composition to the subject.
Parent Case Info

The present application claims the benefit of U.S. Provisional Application No. 63/110,711 filed on Nov. 6, 2020, the entire contents of which are incorporated herein by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2021/058162 11/5/2021 WO
Provisional Applications (1)
Number Date Country
63110711 Nov 2020 US