COMPOSTIONS AND METHODS FOR TREATING PROSTATE CANCER

Information

  • Patent Application
  • 20240009172
  • Publication Number
    20240009172
  • Date Filed
    July 06, 2023
    10 months ago
  • Date Published
    January 11, 2024
    4 months ago
Abstract
The present disclosure relates to compositions, systems, and methods for treating cancer. In particular, the present disclosure relates to compositions, systems, and methods for characterizing and treating prostate cancer (e.g., castration-resistant prostate cancer).
Description
FIELD OF THE DISCLOSURE

The present disclosure relates to compositions, systems, and methods for treating cancer. In particular, the present disclosure relates to compositions, systems, and methods for characterizing and treating prostate cancer (e.g., castration-resistant prostate cancer).


BACKGROUND OF THE DISCLOSURE

Afflicting one out of nine men over age 65, prostate cancer (PCA) is a leading cause of male cancer-related death, second only to lung cancer (Abate-Shen and Shen, Genes Dev 14:2410 [2000]; Ruijter et al., Endocr Rev, 20:22 [1999]). The American Cancer Society estimates that about 184,500 American men will be diagnosed with prostate cancer and 39,200 will die in 2001.


Prostate cancer is typically diagnosed with a digital rectal exam and/or prostate specific antigen (PSA) screening. An elevated serum PSA level can indicate the presence of PCA. PSA is used as a marker for prostate cancer because it is secreted only by prostate cells. A healthy prostate will produce a stable amount—typically below 4 nanograms per milliliter, or a PSA reading of “4” or less—whereas cancer cells produce escalating amounts that correspond with the severity of the cancer. A level between 4 and 10 may raise a doctor's suspicion that a patient has prostate cancer, while amounts above 50 may show that the tumor has spread elsewhere in the body.


When PSA or digital tests indicate a strong likelihood that cancer is present, a transrectal ultrasound (TRUS) is used to map the prostate and show any suspicious areas. Biopsies of various sectors of the prostate are used to determine if prostate cancer is present. Treatment options depend on the stage of the cancer. Men with a 10-year life expectancy or less who have a low Gleason number and whose tumor has not spread beyond the prostate are often treated with watchful waiting (no treatment). Treatment options for more aggressive cancers include surgical treatments such as radical prostatectomy (RP), in which the prostate is completely removed (with or without nerve sparing techniques) and radiation, applied through an external beam that directs the dose to the prostate from outside the body or via low-dose radioactive seeds that are implanted within the prostate to kill cancer cells locally. Anti-androgen hormone therapy is also used, alone or in conjunction with surgery or radiation. Hormone therapy uses luteinizing hormone-releasing hormones (LH-RH) analogs, which block the pituitary from producing hormones that stimulate testosterone production. Patients must have injections of LH-RH analogs for the rest of their lives.


While surgical and hormonal treatments are often effective for localized PCA, advanced disease remains essentially incurable. Androgen ablation is the most common therapy for advanced PCA, leading to massive apoptosis of androgen-dependent malignant cells and temporary tumor regression. In most cases, however, the tumor reemerges with a vengeance and can proliferate independent of androgen signals.


What is needed are improved methods for identifying and treating cancer unlikely to respond to androgen ablation.


SUMMARY OF THE DISCLOSURE

The present disclosure relates to compositions, systems, and methods for treating cancer. In particular, the present disclosure relates to compositions, systems, and methods for characterizing and treating prostate cancer (e.g., castration-resistant prostate cancer).


Experiments described herein identified a gene expression signature that identifies individuals unlikely to respond to androgen deprivation therapy. Such individuals can be offered alternative treatments, thus improving outcomes.


Accordingly, in some embodiments, provided herein is a method for treating prostate cancer, comprising: a) assaying a sample from a subject diagnosed with prostate cancer for the level of expression of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) genes selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1; b) calculating a lineage plasticity score based on the level of gene expression; c) identifying subjects with a high lineage plasticity score; and d) administering a non-androgen receptor signaling inhibitor treatment to the subjects. In some embodiments, a score above 0.577 (e.g., above 0.45, 0.50, 0.55, 0.60, or 0.65) (e.g., as calculated using GSVA), is considered high.


The present disclosure is not limited to particular non-androgen receptor signaling inhibitor treatment. Examples include but are not limited to, chemotherapy, radiation, surgery, or a pharmaceutical agent. In some exemplary embodiments, the treatment is an agent that blocks expression or activity of one or more of the genes. Examples include but are not limited to, an antibody, a nucleic acid, or a small molecule.


Further embodiments provide a method for treating prostate cancer, comprising: a) assaying a sample from a subject diagnosed with prostate cancer for the level of expression of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) genes selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1; b) calculating a lineage plasticity score based on the level of gene expression; c) identifying subjects with a low lineage plasticity score; and d) administering an androgen receptor signaling inhibitor treatment (e.g., enzalutamide) to the subjects.


Additional embodiments provide a method for measuring gene expression, comprising: a) assaying a sample from a subject diagnosed with prostate cancer for the level of expression of two or more genes (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1; b) calculating a lineage plasticity score based on the level of gene expression.


Some embodiments provide a method for measuring gene expression, comprising: assaying a sample from a subject diagnosed with prostate cancer for the level of expression of two or more genes (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1. In some embodiments, the level of expression of no more than 14, 20, 25, 30, 500, or 100 genes are detected. In some embodiments, the level of expression of only RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, and RHOBTB1 is detected.


Yet other embodiments provide a method for providing a prognosis to a subject with prostate cancer, comprising: a) assaying a sample from a subject diagnosed with prostate cancer for the level of expression of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) genes selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1; b) calculating a lineage plasticity score based on the level of gene expression; and c) providing a prognosis of increased likelihood of death when the lineage plasticity score is high.


Still other embodiments provide a method for characterizing prostate cancer a subject with prostate cancer, comprising: a) assaying a sample from a subject diagnosed with prostate cancer for the level of expression of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) genes selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1; b) calculating a lineage plasticity score based on the level of gene expression; and c) providing a prognosis of increased likelihood of said cancer undergoing lineage plasticity when the lineage plasticity score is high.


In some embodiments, the prostate cancer is castration-resistant prostate cancer (CRPC). In some embodiments, the sample is blood, urine or prostate cells.


Also provided is a kit, comprising reagents for detecting the level of expression of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) genes selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1. In some embodiments, the reagents are nucleic acid primers, nucleic acid probes, or antibodies.


Additional embodiments provide a system, comprising: a computer processor and computer software configured to calculate a lineage plasticity score based on the level of expression of one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) genes selected from RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1.


Also provided is the use of an androgen receptor signaling inhibitor to treat prostate cancer in a subject with a low lineage plasticity score.


Additional embodiments will be apparent to persons skilled in the relevant art based on the teachings contained herein.





DESCRIPTION OF THE DRAWINGS


FIG. 1 shows study biopsy and clinical information. a. Study schematic. b. Sankey diagram showing site of biopsy at baseline (left) and at progression (right). c. Left panel shows PSA change at 12 weeks for each patient.



FIG. 2 shows that the effect of enzalutamide on tumor transcriptome is heterogenous across patients. a. Similarity heatmap for all samples clustered by variance-stabilization transformation (vst). b. Clinical and gene expression data for each matched pair ordered on x-axis by time between biopsies.



FIG. 3 shows that pathway and master regulator analysis implicate E2F1 in lineage plasticity risk, and a signature of lineage plasticity risk identifies tumors with poor outcomes after androgen receptor signaling inhibitor treatment. a. Hallmark pathway analysis of activated pathways in baseline samples for the three patients whose tumors converted (underwent lineage plasticity) vs. those patients whose tumors did not upon progression. b. Master regulator analysis identifies top activated and deactivated transcription factors between converters and non-converters using the baseline tumor samples. c. Dot plot showing lineage plasticity signature score for patients in indicated cohorts. d,e. Kaplan-Meier survival curves for patients in the Alumkal, et al. cohort (d) and Abida, et al. cohort (e) stratified by high or low lineage plasticity risk score. f. Dot plot showing lineage plasticity signature score for all castration naïve adenocarcinoma PDX models described by Lin, et al.23



FIG. 4 gene expression profiling and multiplex immunofluorescence that identify gene expression changes in tumors undergoing enzalutamide-induced lineage plasticity. a. Volcano plot showing top up and down regulated genes in progression samples vs. baseline samples for the three patients whose tumors converted. b. ARG10 gene signature heatmap for three converters at baseline and progression. The left half shows the expression levels of individual genes in the ARG10 signature, and the right half shows the ARG10 signature score. p-value shown is for a paired t-test between baseline and progression ARG10 scores (n=3 pairs). c. Hallmark pathway analysis shows the top up or down regulated pathways in progression vs. baseline samples for the three patients whose tumors converted d. Multiplex immunofluorescence for AR, NKX3.1, and HOXB13 expression between baseline vs. progression samples for patient 135, 210, and an additional West Coast Dream Team patient (patient 103) whose tumor converted. Scale bar represents 50 μm.



FIG. 5 shows a. AR VIPER Score for each baseline and progression sample. b. ARG10 and VIPER AR score are strongly correlated. c. AR-V7 splice variant expression for each baseline and progression sample. Signature scores were calculated for baseline and progression samples using Beltran, et al. NEPC upregulated genes in d, Zhang, et al. basal genes in e, Kim, et al. AR-repressed lineage plasticity genes in f, and ARG10 genes in g. h. Unsupervised hierarchical clustering of all baseline samples using top 500 differentially expressed genes. i. Unsupervised hierarchical clustering of all baseline samples using top 1000 differentially expressed genes. j. Signature scores were calculated for baseline and progression samples using genes upregulated with RB1 loss described by Chen, et al.



FIG. 6 shows a. lineage plasticity risk scores calculated for baseline vs. progression samples. b. Dotplot showing lineage plasticity risk signature score for patients described in prostate cancer TCGA15. c. Heatmap showing lineage plasticity risk score in LTL331, other hormone-naïve LTL PDXs, and LTL331R described by Lin, et al. d. Gene set enrichment plot for 14 gene lineage plasticity risk signature in LTL331 vs. other nine hormone-naïve LTL PDXs described in Lin, et al.



FIG. 7 shows Hallmark pathway analysis demonstrating the top up- or downregulated pathways in progression vs. baseline samples for the 18 patients whose tumors did not convert.



FIG. 8 shows a. Panels show expression of AR, NKX3.1, INSM1, and HOXB13 in ARPC LuCaP 96CR PDX tumor, NEPC LuCaP 145.1 PDX tumor, and DNPC LuCaP 173.2 PDX tumor. AR and NKX3.1 were only expressed in LuCaP 96CR. INSM1 was only expressed in LuCaP 145.1, while HOXB13 was expressed in both LuCaP 96CR and LuCaP 173.2. b. Absent INSM1 expression in all three matched converter samples examined.





DEFINITIONS

To facilitate an understanding of the present disclosure, a number of terms and phrases are defined below:


As used herein, the term “sensitivity” is defined as a statistical measure of performance of an assay (e.g., method, test), calculated by dividing the number of true positives by the sum of the true positives and the false negatives.


As used herein, the term “specificity” is defined as a statistical measure of performance of an assay (e.g., method, test), calculated by dividing the number of true negatives by the sum of true negatives and false positives.


As used herein, the term “informative” or “informativeness” refers to a quality of a marker or panel of markers, and specifically to the likelihood of finding a marker (or panel of markers) in a positive sample.


As used herein, the term “metastasis” is meant to refer to the process in which cancer cells originating in one organ or part of the body relocate to another part of the body and continue to replicate. Metastasized cells subsequently form tumors which may further metastasize. Metastasis thus refers to the spread of cancer from the part of the body where it originally occurs to other parts of the body. As used herein, the term “metastasized prostate cancer cells” is meant to refer to prostate cancer cells which have metastasized.


The term “neoplasm” as used herein refers to any new and abnormal growth of tissue. Thus, a neoplasm can be a non-malignant neoplasm, a premalignant neoplasm or a malignant neoplasm. The term “neoplasm-specific marker” refers to any biological material that can be used to indicate the presence of a neoplasm. Examples of biological materials include, without limitation, nucleic acids, polypeptides, carbohydrates, fatty acids, cellular components (e.g., cell membranes and mitochondria), and whole cells.


As used herein, the term “nucleic acid molecule” refers to any nucleic acid containing molecule, including but not limited to, DNA or RNA. The term encompasses sequences that include any of the known base analogs of DNA and RNA including, but not limited to, 4 acetylcytosine, 8-hydroxy-N6-methyladenosine, aziridinylcytosine, pseudoisocytosine, 5-(carboxyhydroxyl-methyl) uracil, 5-fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5-carboxymethyl-aminomethyluracil, dihydrouracil, inosine, N6-isopentenyladenine, 1-methyladenine, 1-methylpseudo-uracil, 1-methylguanine, 1-methylinosine, 2,2-dimethyl-guanine, 2-methyladenine, 2-methylguanine, 3-methyl-cytosine, 5-methylcytosine, N6-methyladenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxy-amino-methyl-2-thiouracil, 0-D-mannosylqueosine, 5′-methoxycarbonylmethyluracil, 5-methoxyuracil, 2-methylthio-N-isopentenyladenine, uracil-acid methylester, uracil-5-oxyacetic acid, oxybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, N-uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, and 2,6-diaminopurine.


As used herein, the term “nucleobase” is synonymous with other terms in use in the art including “nucleotide,” “deoxynucleotide,” “nucleotide residue,” “deoxynucleotide residue,” “nucleotide triphosphate (NTP),” or deoxynucleotide triphosphate (dNTP).


An “oligonucleotide” refers to a nucleic acid that includes at least two nucleic acid monomer units (e.g., nucleotides), typically more than three monomer units, and more typically greater than ten monomer units. The exact size of an oligonucleotide generally depends on various factors, including the ultimate function or use of the oligonucleotide. To further illustrate, oligonucleotides are typically less than 200 residues long (e.g., between 15 and 100), however, as used herein, the term is also intended to encompass longer polynucleotide chains. Oligonucleotides are often referred to by their length. For example, a 24 residue oligonucleotide is referred to as a “24-mer”. Typically, the nucleoside monomers are linked by phosphodiester bonds or analogs thereof, including phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoranilidate, phosphoramidate, and the like, including associated counterions, e.g., 1-1±, NH 4+, Nat, and the like, if such counterions are present. Further, oligonucleotides are typically single-stranded. Oligonucleotides are optionally prepared by any suitable method, including, but not limited to, isolation of an existing or natural sequence, DNA replication or amplification, reverse transcription, cloning and restriction digestion of appropriate sequences, or direct chemical synthesis by a method such as the phosphotriester method of Narang et al. (1979) Meth Enzymol. 68: 90-99; the phosphodiester method of Brown et al. (1979) Meth Enzymol. 68: 109-151; the diethylphosphoramidite method of Beaucage et al. (1981) Tetrahedron Lett. 22: 1859-1862; the triester method of Matteucci et al. (1981) J Am Chem Soc. 103:3185-3191; automated synthesis methods; or the solid support method of U.S. Pat. No. 4,458,066, entitled “PROCESS FOR PREPARING POLYNUCLEOTIDES,” issued Jul. 3, 1984 to Caruthers et al., or other methods known to those skilled in the art. All of these references are incorporated by reference.


A “sequence” of a biopolymer refers to the order and identity of monomer units (e.g., nucleotides, etc.) in the biopolymer. The sequence (e.g., base sequence) of a nucleic acid is typically read in the 5′ to 3′ direction.


As used herein, the term “subject” refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, rodents, and the like, which is to be the recipient of a particular treatment. Typically, the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.


As used herein, the term “non-human animals” refers to all non-human animals including, but are not limited to, vertebrates such as rodents, non-human primates, ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, canines, felines, ayes, etc.


As used herein, the term “sample” is used in its broadest sense. In one sense, it is meant to include a specimen or culture obtained from any source, as well as biological and environmental samples. Biological samples may be obtained from animals (including humans) and encompass fluids, solids, tissues, and gases. Biological samples include blood products, such as plasma, serum and the like. Such examples are not however to be construed as limiting the sample types applicable to the present invention.


DETAILED DESCRIPTION OF THE DISCLOSURE

The present disclosure relates to compositions, systems, and methods for treating cancer. In particular, the present disclosure relates to compositions, systems, and methods for characterizing and treating prostate cancer (e.g., castration-resistant prostate cancer).


Androgen deprivation therapy (ADT) is the principal treatment for metastatic prostate cancer, but progression to castration-resistant prostate cancer (CRPC) is nearly universal. In recent years, potent inhibitors of the androgen receptor (AR)—a luminal lineage transcription factor—have been developed, including the AR antagonist enzalutamide (enza) 1-5. Enza improves progression-free survival and overall survival in patients with CRPC; further, enza also increases overall survival in patients with hormone-naïve prostate cancer who are beginning ADT for the first time 6-9. However, one-third of patients do not respond, and those with de novo resistance have a significantly increased risk of death compared to responders 6-9.


Despite intense study, clinical enza resistance remains poorly understood. Several studies examined mechanisms of de novo or acquired enza resistance in clinical samples and implicated: AR amplification,10,11 AR splice variants,12,13 increased Wnt/r3-catenin signaling,14-16 increased TGF-β signaling,15,17 epithelial to mesenchymal transition or increased stemness,15,18 and lineage plasticity 15. However, these prior studies were largely restricted to DNA mutational profiling, compared baseline and progression samples from different patients, used limited numbers of matched samples, or did not focus on transcriptional changes.


Reports have indicated that most CRPC tumors resistant to AR signaling inhibitors (ARSIs) continue to depend on the AR 18,19. However, lineage plasticity 20—most commonly exemplified by loss of AR signaling and a switch from a luminal to an alternate differentiation program—is a resistance mechanism that appears to be increasing in the era of more widespread use of ARSIs. The emergence of tumors with features of lineage plasticity may occur through diverse mechanisms: selection of a pre-existing clone that has already undergone differentiation change, acquisition of new genetic alterations that promote differentiation change, or transdifferentiation of tumor cells through epigenetic mechanisms 18, 21-23.


Lineage plasticity is a continuum, ranging from tumors with persistent AR expression but low AR activity, those that lose AR expression but do not undergone neuroendocrine differentiation (double negative prostate cancer (DNPC)), and those that lose AR expression and do undergo neuroendocrine differentiation (neuroendocrine prostate cancer (NEPC) 24. Importantly, CRPC tumors that have undergone lineage plasticity are associated with a much shorter survival than CRPC tumors that have persistent AR activity and a luminal lineage program, demonstrating an urgent need to understand treatment-induced lineage plasticity in prostate cancer 25.


Experiments described herein compared gene expression profiles between matched CRPC tumor biopsy samples prior to enza and at the time of progression to identify pre-treatment and treatment-induced resistance mechanisms in individual patients. Results from 21 matched samples demonstrated key transcriptional differences, including lineage plasticity changes induced by enza, that contribute to resistance.


Accordingly, provided herein are compositions and methods for characterizing and treating prostate cancer. In some embodiments, the compositions and methods of the present disclosure utilize a 14 gene signature of lineage plasticity to identify subjects most likely to benefit from AR targeted therapy. In some embodiments, the level of expression of the lineage plasticity signature (e.g., one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all) of RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, or RHOBTB1 is utilized to calculate a lineage plasticity score.


In some embodiments, lineage plasticity scores are calculated using gene expression data. In some embodiments, the single-sample gene set enrichment analysis (ssGSEA) 8 implemented in the GSVA 9 R package is used to calculate the score.


In some embodiments, a numerical cut-off for a “high” lineage plasticity score is utilized. For example, in some embodiments, a score above 0.577 (e.g., above 0.45, 0.50, 0.60, or 0.65) (e.g., as calculated using GSVA or other method), is considered high. The present invention is not limited to particular methods of detecting the level of the recited markers. Markers may be detected as DNA (e.g., cDNA), RNA (e.g., mRNA), or protein.


In some embodiments, nucleic acid sequencing methods are utilized for detection. In some embodiments, the technology provided herein finds use in a Second Generation (a.k.a. Next Generation or Next-Gen), Third Generation (a.k.a. Next-Next-Gen), or Fourth Generation (a.k.a. N3-Gen) sequencing technology including, but not limited to, pyrosequencing, sequencing-by-ligation, single molecule sequencing, sequence-by-synthesis (SBS), semiconductor sequencing, massive parallel clonal, massive parallel single molecule SBS, massive parallel single molecule real-time, massive parallel single molecule real-time nanopore technology, etc. Morozova and Marra provide a review of some such technologies in Genomics, 92: 255 (2008), herein incorporated by reference in its entirety. Those of ordinary skill in the art will recognize that because RNA is less stable in the cell and more prone to nuclease attack experimentally RNA is usually reverse transcribed to DNA before sequencing.


A number of DNA sequencing techniques are suitable, including fluorescence-based sequencing methodologies (See, e.g., Birren et al., Genome Analysis: Analyzing DNA, 1, Cold Spring Harbor, N.Y.; herein incorporated by reference in its entirety). In some embodiments, the technology finds use in automated sequencing techniques understood in that art. In some embodiments, the present technology finds use in parallel sequencing of partitioned amplicons (PCT Publication No: WO2006084132 to Kevin McKernan et al., herein incorporated by reference in its entirety). In some embodiments, the technology finds use in DNA sequencing by parallel oligonucleotide extension (See, e.g., U.S. Pat. No. 5,750,341 to Macevicz et al., and U.S. Pat. No. 6,306,597 to Macevicz et al., both of which are herein incorporated by reference in their entireties). Additional examples of sequencing techniques in which the technology finds use include the Church polony technology (Mitra et al., 2003, Analytical Biochemistry 320, 55-65; Shendure et al., 2005 Science 309, 1728-1732; U.S. Pat. Nos. 6,432,360, 6,485,944, 6,511,803; herein incorporated by reference in their entireties), the 454 picotiter pyrosequencing technology (Margulies et al., 2005 Nature 437, 376-380; US 20050130173; herein incorporated by reference in their entireties), the Solexa single base addition technology (Bennett et al., 2005, Pharmacogenomics, 6, 373-382; U.S. Pat. Nos. 6,787,308; 6,833,246; herein incorporated by reference in their entireties), the Lynx massively parallel signature sequencing technology (Brenner et al. (2000). Nat. Biotechnol. 18:630-634; U.S. Pat. Nos. 5,695,934; 5,714,330; herein incorporated by reference in their entireties), and the Adessi PCR colony technology (Adessi et al. (2000). Nucleic Acid Res. 28, E87; WO 00018957; herein incorporated by reference in its entirety).


Next-generation sequencing (NGS) methods share the common feature of massively parallel, high-throughput strategies, with the goal of lower costs in comparison to older sequencing methods (see, e.g., Voelkerding et al., Clinical Chem., 55: 641-658, 2009; MacLean et al., Nature Rev. Microbiol., 7: 287-296; each herein incorporated by reference in their entirety). NGS methods can be broadly divided into those that typically use template amplification and those that do not Amplification-requiring methods include pyrosequencing commercialized by Roche as the 454 technology platforms (e.g., GS 20 and GS FLX), Life Technologies/Ion Torrent, the Solexa platform commercialized by Illumina, GnuBio, and the Supported Oligonucleotide Ligation and Detection (SOLiD) platform commercialized by Applied Biosystems. Non-amplification approaches, also known as single-molecule sequencing, are exemplified by the HeliScope platform commercialized by Helicos BioSciences, and emerging platforms commercialized by VisiGen, Oxford Nanopore Technologies Ltd., and Pacific Biosciences, respectively.


In some embodiments, hybridization methods are utilized. Illustrative non-limiting examples of nucleic acid hybridization techniques include, but are not limited to, in situ hybridization (ISH), microarray, and Southern or Northern blot.


In situ hybridization (ISH) is a type of hybridization that uses a labeled complementary DNA or RNA strand as a probe to localize a specific DNA or RNA sequence in a portion or section of tissue (in situ), or, if the tissue is small enough, the entire tissue (whole mount ISH). DNA ISH can be used to determine the structure of chromosomes. RNA ISH is used to measure and localize mRNAs and other transcripts within tissue sections or whole mounts. Sample cells and tissues are usually treated to fix the target transcripts in place and to increase access of the probe. The probe hybridizes to the target sequence at elevated temperature, and then the excess probe is washed away. The probe that was labeled with radio-, fluorescent- or antigen-labeled bases is localized and quantitated in the tissue using autoradiography, fluorescence microscopy or immunohistochemistry. ISH can also use two or more probes, labeled with radioactivity or the other non-radioactive labels, to simultaneously detect two or more transcripts.


In some embodiments, markers are detected using fluorescence in situ hybridization (FISH). The preferred FISH assays for methods of embodiments of the present disclosure utilize bacterial artificial chromosomes (BACs). These have been used extensively in the human genome sequencing project (see Nature 409: 953-958 (2001)) and clones containing specific BACs are available through distributors that can be located through many sources, e.g., NCBI. Each BAC clone from the human genome has been given a reference name that unambiguously identifies it. These names can be used to find a corresponding GenBank sequence and to order copies of the clone from a distributor.


Different kinds of biological assays are called microarrays including, but not limited to: microarrays (e.g., cDNA microarrays and oligonucleotide microarrays); protein microarrays; tissue microarrays; transfection or cell microarrays; chemical compound microarrays; and, antibody microarrays. A DNA microarray, commonly known as gene chip, DNA chip, or biochip, is a collection of microscopic DNA spots attached to a solid surface (e.g., glass, plastic or silicon chip) forming an array for the purpose of expression profiling or monitoring expression levels for thousands of genes simultaneously. The affixed DNA segments are known as probes, thousands of which can be used in a single DNA microarray. Microarrays can be used to identify disease genes by comparing gene expression in disease and normal cells. Microarrays can be fabricated using a variety of technologies, including but not limited to: printing with fine-pointed pins onto glass slides; photolithography using pre-made masks; photolithography using dynamic micromirror devices; ink-jet printing; or, electrochemistry on microelectrode arrays.


Southern and Northern blotting may be used to detect specific DNA or RNA sequences, respectively. In these techniques DNA or RNA is extracted from a sample, fragmented, electrophoretically separated on a matrix gel, and transferred to a membrane filter. The filter bound DNA or RNA is subject to hybridization with a labeled probe complementary to the sequence of interest. Hybridized probe bound to the filter is detected. A variant of the procedure is the reverse Northern blot, in which the substrate nucleic acid that is affixed to the membrane is a collection of isolated DNA fragments and the probe is RNA extracted from a tissue and labeled.


In some embodiments, marker sequences are amplified (e.g., after conversion to DNA) prior to or simultaneous with detection. Illustrative non-limiting examples of nucleic acid amplification techniques include, but are not limited to, polymerase chain reaction (PCR), reverse transcription polymerase chain reaction (RT-PCR), transcription-mediated amplification (TMA), ligase chain reaction (LCR), strand displacement amplification (SDA), and nucleic acid sequence based amplification (NASBA). Those of ordinary skill in the art will recognize that certain amplification techniques (e.g., PCR) require that RNA be reversed transcribed to DNA prior to amplification (e.g., RT-PCR), whereas other amplification techniques directly amplify RNA (e.g., TMA and NASBA).


In some embodiments, quantitative evaluation of the amplification process in real-time is performed. Evaluation of an amplification process in “real-time” involves determining the amount of amplicon in the reaction mixture either continuously or periodically during the amplification reaction, and using the determined values to calculate the amount of target sequence initially present in the sample. A variety of methods for determining the amount of initial target sequence present in a sample based on real-time amplification are well known in the art. These include methods disclosed in U.S. Pat. Nos. 6,303,305 and 6,541,205, each of which is herein incorporated by reference in its entirety. Another method for determining the quantity of target sequence initially present in a sample, but which is not based on a real-time amplification, is disclosed in U.S. Pat. No. 5,710,029, herein incorporated by reference in its entirety.


Amplification products may be detected in real-time through the use of various self-hybridizing probes, most of which have a stem-loop structure. Such self-hybridizing probes are labeled so that they emit differently detectable signals, depending on whether the probes are in a self-hybridized state or an altered state through hybridization to a target sequence. By way of non-limiting example, “molecular torches” are a type of self-hybridizing probe that includes distinct regions of self-complementarity (referred to as “the target binding domain” and “the target closing domain”) which are connected by a joining region (e.g., non-nucleotide linker) and which hybridize to each other under predetermined hybridization assay conditions. In a preferred embodiment, molecular torches contain single-stranded base regions in the target binding domain that are from 1 to about 20 bases in length and are accessible for hybridization to a target sequence present in an amplification reaction under strand displacement conditions. Under strand displacement conditions, hybridization of the two complementary regions, which may be fully or partially complementary, of the molecular torch is favored, except in the presence of the target sequence, which will bind to the single-stranded region present in the target binding domain and displace all or a portion of the target closing domain. The target binding domain and the target closing domain of a molecular torch include a detectable label or a pair of interacting labels (e.g., luminescent/quencher) positioned so that a different signal is produced when the molecular torch is self-hybridized than when the molecular torch is hybridized to the target sequence, thereby permitting detection of probe:target duplexes in a test sample in the presence of unhybridized molecular torches. Molecular torches and a variety of types of interacting label pairs, including fluorescence resonance energy transfer (FRET) labels, are disclosed in, for example U.S. Pat. Nos. 6,534,274 and 5,776,782, each of which is herein incorporated by reference in its entirety.


Another example of a detection probe having self-complementarity is a “molecular beacon.” Molecular beacons include nucleic acid molecules having a target complementary sequence, an affinity pair (or nucleic acid arms) holding the probe in a closed conformation in the absence of a target sequence present in an amplification reaction, and a label pair that interacts when the probe is in a closed conformation. Hybridization of the target sequence and the target complementary sequence separates the members of the affinity pair, thereby shifting the probe to an open conformation. The shift to the open conformation is detectable due to reduced interaction of the label pair, which may be, for example, a fluorophore and a quencher (e.g., DABCYL and EDANS). Molecular beacons are disclosed, for example, in U.S. Pat. Nos. 5,925,517 and 6,150,097, herein incorporated by reference in its entirety.


The cancer marker genes described herein may be detected as proteins using a variety of protein techniques known to those of ordinary skill in the art, including but not limited to: protein sequencing; and, immunoassays.


Illustrative non-limiting examples of protein sequencing techniques include, but are not limited to, mass spectrometry and Edman degradation.


Mass spectrometry can, in principle, sequence any size protein but becomes computationally more difficult as size increases. A protein is digested by an endoprotease, and the resulting solution is passed through a high pressure liquid chromatography column. At the end of this column, the solution is sprayed out of a narrow nozzle charged to a high positive potential into the mass spectrometer. The charge on the droplets causes them to fragment until only single ions remain. The peptides are then fragmented and the mass-charge ratios of the fragments measured. The mass spectrum is analyzed by computer and often compared against a database of previously sequenced proteins in order to determine the sequences of the fragments. The process is then repeated with a different digestion enzyme, and the overlaps in sequences are used to construct a sequence for the protein.


In the Edman degradation reaction, the peptide to be sequenced is adsorbed onto a solid surface (e.g., a glass fiber coated with polybrene). The Edman reagent, phenylisothiocyanate (PTC), is added to the adsorbed peptide, together with a mildly basic buffer solution of 12% trimethylamine, and reacts with the amine group of the N-terminal amino acid. The terminal amino acid derivative can then be selectively detached by the addition of anhydrous acid. The derivative isomerizes to give a substituted phenylthiohydantoin, which can be washed off and identified by chromatography, and the cycle can be repeated. The efficiency of each step is about 98%, which allows about 50 amino acids to be reliably determined.


Illustrative non-limiting examples of immunoassays include, but are not limited to: immunoprecipitation; Western blot; ELISA; immunohistochemistry; immunocytochemistry; flow cytometry; and, immuno-PCR. Polyclonal or monoclonal antibodies detectably labeled using various techniques known to those of ordinary skill in the art (e.g., colorimetric, fluorescent, chemiluminescent or radioactive) are suitable for use in the immunoassays. Immunoprecipitation is the technique of precipitating an antigen out of solution using an antibody specific to that antigen. The process can be used to identify protein complexes present in cell extracts by targeting a protein believed to be in the complex. The complexes are brought out of solution by insoluble antibody-binding proteins isolated initially from bacteria, such as Protein A and Protein G. The antibodies can also be coupled to sepharose beads that can easily be isolated out of solution. After washing, the precipitate can be analyzed using mass spectrometry, Western blotting, or any number of other methods for identifying constituents in the complex.


A Western blot, or immunoblot, is a method to detect protein in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate denatured proteins by mass. The proteins are then transferred out of the gel and onto a membrane, typically polyvinyldiflroride or nitrocellulose, where they are probed using antibodies specific to the protein of interest. As a result, researchers can examine the amount of protein in a given sample and compare levels between several groups.


An ELISA, short for Enzyme-Linked ImmunoSorbent Assay, is a biochemical technique to detect the presence of an antibody or an antigen in a sample. It utilizes a minimum of two antibodies, one of which is specific to the antigen and the other of which is coupled to an enzyme. The second antibody will cause a chromogenic or fluorogenic substrate to produce a signal. Variations of ELISA include sandwich ELISA, competitive ELISA, and ELISPOT. Because the ELISA can be performed to evaluate either the presence of antigen or the presence of antibody in a sample, it is a useful tool both for determining serum antibody concentrations and also for detecting the presence of antigen.


Immunohistochemistry and immunocytochemistry refer to the process of localizing proteins in a tissue section or cell, respectively, via the principle of antigens in tissue or cells binding to their respective antibodies. Visualization is enabled by tagging the antibody with color producing or fluorescent tags. Typical examples of color tags include, but are not limited to, horseradish peroxidase and alkaline phosphatase. Typical examples of fluorophore tags include, but are not limited to, fluorescein isothiocyanate (FITC) or phycoerythrin (PE).


Immuno-polymerase chain reaction (IPCR) utilizes nucleic acid amplification techniques to increase signal generation in antibody-based immunoassays. Because no protein equivalence of PCR exists, that is, proteins cannot be replicated in the same manner that nucleic acid is replicated during PCR, the only way to increase detection sensitivity is by signal amplification. The target proteins are bound to antibodies which are directly or indirectly conjugated to oligonucleotides. Unbound antibodies are washed away and the remaining bound antibodies have their oligonucleotides amplified. Protein detection occurs via detection of amplified oligonucleotides using standard nucleic acid detection methods, including real-time methods.


Embodiments of the present invention further provide kits and systems comprising reagents for detection of the recited markers (e.g., primer, probes, etc.). In some embodiments, kits and systems comprise computer systems for analyzing marker levels and providing a lineage plasticity score, diagnoses, prognoses, or determining treatment courses of action.


In some embodiments, a computer-based analysis program is used to translate the raw data generated by the detection assay (e.g., levels of the recited markers) into data of predictive value for a clinician. The clinician can access the predictive data using any suitable means. Thus, in some preferred embodiments, the present invention provides the further benefit that the clinician, who is not likely to be trained in genetics or molecular biology, need not understand the raw data. The data is presented directly to the clinician in its most useful form. The clinician is then able to immediately utilize the information in order to optimize the care of the subject.


The present invention contemplates any method capable of receiving, processing, and transmitting the information to and from laboratories conducting the assays, information provides, medical personal, and subjects. For example, in some embodiments of the present invention, a sample (e.g., a biopsy or a serum or urine sample) is obtained from a subject and submitted to a profiling service (e.g., clinical lab at a medical facility, genomic profiling business, etc.), located in any part of the world (e.g., in a country different than the country where the subject resides or where the information is ultimately used) to generate raw data. Where the sample comprises a tissue or other biological sample, the subject may visit a medical center to have the sample obtained and sent to the profiling center, or subjects may collect the sample themselves (e.g., a urine or blood sample) and directly send it to a profiling center. Where the sample comprises previously determined biological information, the information may be directly sent to the profiling service by the subject (e.g., an information card containing the information may be scanned by a computer and the data transmitted to a computer of the profiling center using an electronic communication system). Once received by the profiling service, the sample is processed and a profile is produced (i.e., marker levels) specific for the diagnostic or prognostic information desired for the subject.


The profile data is then prepared in a format suitable for interpretation by a treating clinician. For example, rather than providing raw data, the prepared format may represent a diagnosis or risk assessment (e.g., level of markers) for the subject, along with recommendations for particular treatment options. The data may be displayed to the clinician by any suitable method. For example, in some embodiments, the profiling service generates a report that can be printed for the clinician (e.g., at the point of care) or displayed to the clinician on a computer monitor.


In some embodiments, the information is first analyzed at the point of care or at a regional facility. The raw data is then sent to a central processing facility for further analysis and/or to convert the raw data to information useful for a clinician or patient. The central processing facility provides the advantage of privacy (all data is stored in a central facility with uniform security protocols), speed, and uniformity of data analysis. The central processing facility can then control the fate of the data following treatment of the subject. For example, using an electronic communication system, the central facility can provide data to the clinician, the subject, or researchers.


In some embodiments, the subject is able to directly access the data using the electronic communication system. The subject may chose further intervention or counseling based on the results. In some embodiments, the data is used for research. For example, the data may be used to further optimize the inclusion or elimination of markers as useful indicators of a particular condition or stage of disease or as a companion diagnostic to determine a treatment course of action.


In some specific embodiments, the lineage plasticity score described herein finds use in characterizing, prognosing, and treating prostate cancer. For example, in some embodiments, the score is used to identify individuals likely to develop lineage plasticity (e.g., individuals with a high lineage plasticity score) and corresponding resistance to AR blocking therapy such as enza. Such individuals are offered alternative therapies (e.g., surgery, radiation, chemotherapy, immune therapy, or agents targeted to the genes in the lineage plasticity signature).


Conversely, individuals with a low lineage plasticity score are likely to respond to AR blocking therapy and are thus offered an AR blocking therapy such as enza or other hormone therapy.


Additional hormonal therapies include but are not limited to, leuprolide, goserelin, triptorelin, leuprolide mesylate, degarelix, relugolix, abiraterone, ketoconazole, flutamide, bicalutamide, nilutamide, apalutamide, and darolutamide.


Examples of chemotherapy used in prostate cancer include but are not limited to, docetaxel, cabazitaxel, mitoxantrone, and estramustine. Examples of immnotherapy used in prostate cancer include but are not limited to, cancer vaccines (e.g., sipuleucel-T) and immune checkpoint inhibitors (e.g., pembrolizumab). Additional prostate cancer treatments include but are not limited to, PARP inhibitors (e.g., rucaparib and olaparib).


In some embodiments, a high lineage plasticity score is indicative of an individual with an increased likelihood of death from prostate cancer. In some embodiments, such individuals are offered more aggressive treatments.


As described above, in some embodiments, the present disclosure provides agents that target (e.g., inhibit the expression or one or more activities of) a gene in a lineage plasticity signature. Examples include but are not limited to, small molecules, nucleic acids, and antibodies.


In some embodiments, the inhibitor is a nucleic acid. Exemplary nucleic acids suitable for inhibiting expression of the described markers (e.g., by preventing expression of the marker) include, but are not limited to, antisense nucleic acids and RNAi. In some embodiments, nucleic acid therapies are complementary to and hybridize to at least a portion (e.g., at least 5, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides) of a marker described herein.


In some embodiments, compositions comprising oligomeric antisense compounds, particularly oligonucleotides are used to modulate the function of nucleic acid molecules encoding a marker described herein, ultimately modulating the amount of marker gene expressed. This is accomplished by providing antisense compounds that specifically hybridize with one or more nucleic acids encoding the marker genes. The specific hybridization of an oligomeric compound with its target nucleic acid interferes with the normal function of the nucleic acid. This modulation of function of a target nucleic acid by compounds that specifically hybridize to it is generally referred to as “antisense.” The functions of DNA to be interfered with include replication and transcription. The functions of RNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity that may be engaged in or facilitated by the RNA. The overall effect of such interference with target nucleic acid function is decreasing the amount of marker expressed.


The present disclosure further provides pharmaceutical compositions (e.g., comprising the compounds described above). The pharmaceutical compositions of the present disclosure may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic and to mucous membranes including vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.


In some embodiments, one or more targeted therapies are administered in combination with an existing therapy for prostate cancer.


In some embodiments, agents described herein are screening for activity against prostate cancer (e.g., in vitro drug screening assays or in a clinical study).


EXPERIMENTAL

The following examples are provided in order to demonstrate and further illustrate certain preferred embodiments and aspects of the present disclosure and are not to be construed as limiting the scope thereof.


Example 1
Methods

West Coast Dream Team (WCDT) Metastatic Tissue Collection


Methods for tissue collection have been described previously 48. RNA-sequencing was performed on matched, paired biopsies from 21 men with metastatic, castration-resistant prostate cancer who had a tissue biopsy performed prior to starting treatment with enza and a second biopsy performed at time of progression.


RNA-Sequencing and Data Processing


Core biopsy samples were flash frozen in Optical Cutting Temperature (OCT) for gene expression analysis. Laser capture microdissection was performed on frozen sections to enrich for tumor content 49. Total RNA was isolated (Stratagene Absolutely RNA Nano Prep) (RIN>8) and amplified using NuGEN Ovation RNA seq System V2. Libraries were generated using NuGEN Ovation Ultralow System V2 for Illumina sequencing. RNA seq was performed on the Illumina NextSeq 500, PE75 with at least 100M read pairs. The raw fastq files were first quality checked using FastQC (version 0.11.8) software (http://www.bioinformatics.bbsrc.ac.uk/projects/fastqc/). Fastq files were aligned to hg38 human reference genome and per-gene counts and transcripts per million (TPM) quantified by RSEM 50 (version 1.3.1) based on the gene annotation gencode.v28.annotation.gtf.


Unsupervised Clustering


To understand the overall transcriptional similarities across these 21 paired samples, unsupervised clustering was performed using RNA-sequencing data. Briefly, the raw count matrix was filtered to remove low expression genes and genes with raw count >=20 in at least two samples were kept. The filtered count matrix was transformed using the vst function implemented in DESeq2 R package (version 1.22.2) 51. The transformed values were used to compute the sample-to-sample Euclidean distance metric for hierarchical clustering through the ‘complete’ method. To cluster samples prior to treatment (baseline), TPM gene expression data was first filtered to remove low expression genes as described above and non-protein-coding genes as annotated by HUGO Gene Nomenclature Committee (HGNC). The filtered TPM matrix was log transformed and the 500 most varying genes were selected to compute the sample-to-sample gene expression spearman correlation which was then converted to distance followed by clustering through the ‘complete-linkage hierarchical clustering’ method.


Differential expression gene, pathway, and master regulator analysis Differential gene expression analysis was performed using DESeq2 (version 1.22.2). Gene expression differences were considered significant if passing the following criteria: adjusted P-value <0.05, absolute fold change >1.5. For the converter vs non-converter baseline sample comparison, we used the adjusted P-value <0.1. The Wald test statistics from DESeq2 output was used as pre-ranked gene list scores to perform pathway analysis using cameraPR implemented in limma R package (version 3.38.3) 52 and the hallmark collection from MSigDB database (version 7.0). Transcription factor activity was inferred using the master regulator inference algorithm 53 (MARINa) implemented in the viper R package (version 1.16.0) 26. Pre-ranked gene list scores and a regulatory network (regulome) are the two sources of data required as input for viper analysis. The pre-ranked gene list scores were the same as above and the transcription factor regulome used in this study was curated from several databases as previously described 54.


Single Sample AR Activity


To measure single-sample AR regulon activity, the viper R package (version 1.16.0) 26 with the log2 transformed TPM gene expression matrix as input was used. The regulon used in viper analysis was the same as described above. Scores were considered to have marked difference if change between baseline and progression sample was >20% of the range between all samples.


Multiplex Immunofluorescence


Multiplex immunofluorescence studies using AR- (Cell signaling Technologies, 5153T), INSM1- (Santa Cruz, sc-271408), NKX3.1- (Fisher, 5082788) and HOXB13- (Cell signaling Technologies, 90944S) specific antibodies were carried out on archival formalin fixed paraffin embedded (FFPE) tissues. In brief, 5 μM paraffin sections were de-waxed and rehydrated following standard protocols. The staining protocol consisted of four sequential staining steps, each with tyramide-based signal amplification using the Tyramide SuperBoost kits (Thermo Fisher) as described previously 55. De-waxed slides were first subjected to steaming for 40 min in Target Retrieval Solution (S1700, Agilent) and incubated with AR specific antibodies (1:00). Signal amplification was carried out by first incubating slides with PowerVision Poly-AP Anti-Rabbit (Leica) secondary antibodies followed by Tyramide568 (Tyramide SuperBoost kit, Thermo Fisher) according to manufacturer's protocols. Slides were then stripped by steaming in citrate buffer (Vector) for 20 minutes and subsequently incubated with INSM1 specific antibodies (1:50) followed by PowerVision Poly-AP Anti-mouse (Leica) secondary antibodies and Tyramide647 (Tyramide SuperBoost kit). Next, slides were stripped for 20 minutes in Target Retrieval Solution (S1700, Agilent), incubated with NKX3.1 specific antibodies (1:200) followed by PowerVision Poly-AP Anti-rabbit (Leica) secondary antibodies and Tyramide488 (Tyramide SuperBoost kit). Lastly, slides were steamed in in Citrate buffer (Vector) for 20 minutes, incubated with HOXB13 antibodies (1:50) followed by PowerVision Poly-AP Anti-rabbit (Leica) secondary antibodies and Tyramide350 (Tyramide SuperBoost kit). Slides were mounted with Prolong (Thermo Fisher), imaged on a Nikon Eclipse E800 (Nikon) microscope and image analyses were carried out using QuPath (v0.3.0) 56.


DNA-Sequencing


Next generation targeted genomic DNA-sequencing of FFPE tissue was performed using a 124 gene as previously described 57. Cell-free DNA was extracted from approximately 1 mL of previously banked plasma and subjected to low-pass whole-genome-sequencing (WGS) and targeted deep sequencing using the Ion Torrent™ Next-Generation Sequencing (NGS) system (Thermo Fisher Scientific, Waltham, MA), as described previously 58. NGS reads were processed using Ion Torrent Suite™ and analyzed with standard workflows in Ion Reporter™ (Thermo Fisher Scientific) and established in-house bioinformatics pipelines. Tumor content estimates were derived from low-pass WGS data using the ichorCNA package in R 59. Total mapped NGS reads for low-pass WGS ranged from 4,235,342-6,185,948 (corresponding to 0.202-0.292× coverage). Targeted deep sequencing was performed using the Oncomine™ Comprehensive Assay Plus (Thermo Fisher Scientific), which targets greater than 1 Mb of genomic sequence corresponding to more than 500 genes recurrently altered in human cancers; total mapped NGS reads for targeted sequencing ranged from 5,069,230-8,497,096 (corresponding to 347-596× coverage across the targeted regions). Prioritized variants and copy number alterations from targeted NGS data were manually curated by an experienced molecular pathologist (A.M.U.) using previously established criteria.60


Aggarwal, et al. Cluster Designation


The unsupervised analysis from Aggarwal, et al. 25 identified five clusters using 119 samples. That study identified 528 genes that were the most differentially expressed between the clusters. Using that gene list, cluster assignments for new samples included in this matched biopsy cohort were determined without replicating the unsupervised analysis. First, the sample batch effect between the samples from the previous study and those from the current study was addressed with exponential normalization on the expression data of all samples—old and new. Exponential normalization is a per-sample operation that fits the expression of all genes to a unit exponential distribution. Next, scikit-learn's k-nearest-neighbor classifier implementation (Pedregosa, F., et al. Scikit-learn: Machine Learning in Python. J Mach Learn Res 12, 2825-2830 (2011)) was used to train a classification model using 118 exponential-normalized samples that had pre-existing cluster assignments. The model used 507 genes from the 528-gene list from Aggarwal, et al. 25 because several genes were not expressed in the previously uncharacterized samples used in this report. The model's accuracy in leave-one-out cross validation was 0.712. The trained model was then used to predict the cluster assignment of previously unclassified, exponential-normalized samples.


Labrecque, et al. Classification


To determine the Labrecque classification, a 26 gene signature used previously to define five phenotypic categories of CRPC 24 was applied: AR-high prostate cancer (ARPC), amphicrine prostate cancer, AR-low prostate cancer (ARLPC), double-negative prostate cancer (DNPC), and neuroendocrine prostate cancer (NEPC) 24. One gene (TARP) was missing from the dataset and was not included. Samples were assigned to the phenotype groups by clustering using Euclidean distance calculated by the dist function and visualization using classical multidimensional scaling (MDS) calculated with the cmdscale function in R using the log2(TPM+1) transformed expression profiles of the remaining 25 genes.


Single-Sample Gene Signature Scores


In this study, several gene signatures collected from public resources, including Zhang Basal gene signature 28, Beltran, et al. NEPC Up gene signature 22, ARG10 signature 27, and Kim, et al. 76 gene AR-repressed signature 29 were used. The signature genes are listed in Table 8. TPM gene expression values were log2(TPM+1) transformed and converted to z-scores by: z=(x−μ)/σ, where μ is the average log2(TPM+1) across all samples of a gene and 6 is the standard deviation of the log2(TPM+1) across all samples of a gene. The signature score of each sample was the average z-score of all genes in each signature.


Development of a Lineage Plasticity Risk Gene Signature


To derive the lineage plasticity risk signature, differential gene expression analysis was performed using DESeq2 as described above by comparing baseline converter vs. non-converter samples. Genes upregulated in converter samples with adjusted P value <0.1 were included (Table 5). Single-sample lineage plasticity risk signature was derived using the single-sample gene set enrichment analysis (ssGSEA) 8 implemented in the GSVA 9 R package.


Assessment of the Lineage Plasticity Signature in Patient-Derived Xenograft Models


Baseline gene expression was examined from 10 human prostate adenocarcinoma PDX models 23. Gene expression of the one tumor (LTL331) that undergoes castration-induced lineage plasticity vs. those that do not were compared: LTL310, LTL311, LTL412, LTL-418, LTL313A, LTL313B, LTL313C, LTL313D, and LTL313H. Then, the fold-change-based gene ranking from the comparison was used to assess the enrichment of the lineage plasticity risk signature we identified using gene set enrichment analysis (Subramanian, A., et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 102, 15545-15550 (2005)).


Survival Analysis


Correlation of the lineage plasticity risk signature with survival time was evaluated in two independent datasets. First, after excluding patients that overlapped with this current study, 17 patients whose tumors had undergone RNA-seq from the prior prospective enza clinical trial with overall survival information were identified 18. Second, samples from the International Dream Team dataset for which overall survival from first line ARSI treatment was available were identified; patients were restricted to those without prior exposure to abiraterone, enza or docetaxel 10. Then, the gene expression of the three datasets, including the samples in the matched biopsy cohort, was merged into one matrix to calculate the enrichment score of each sample consistently. Single-sample lineage plasticity risk score was derived using the single-sample gene set enrichment analysis (ssGSEA) (Barbie, D. A., et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108-112 (2009)) implemented in the GSVA R package (Hanzelmann, S, Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013)). A signature cutoff was defined to separate the baseline converter samples from the non-converter samples from the matched biopsy cohort with the maximum margin as calculated by taking the average of the lowest score in the non-convert group and highest score in the converter group. Finally, this cutoff was used to stratify samples in the two independent datasets into two groups with high and lineage plasticity signature risk scores. The comparison of the survival pattern between the two groups was performed by the Kaplan-Meier method using the Mantel-Cox log-rank test.


SU2C Sample Relabeling


For several samples, aSU2C IDs were relabeled as baseline or progression based upon when the patient was exposed to enzalutamide. DTB_022_PRO and DTB_024_PRO were relabeled DTB_022_BL and DTB_024_BL, respectively, as those biopsies were performed immediately prior to starting enzalutamide treatment. Correspondingly, DTB_022_PRO2 and DTB_024_PRO2 were relabeled DTB_022_PRO and DTB_024_PRO as those biopsies were performed at progression on enzalutamide. DTB_089_PRO2 was relabeled DTB_089_PRO as patient continued enzalutamide until just after PRO2 biopsy.


Results

By examining the Stand Up to Cancer Foundation/Prostate Cancer Foundation West Coast Dream Team (WCDT) prospective cohort, 21 patients with CRPC who underwent a metastatic tumor biopsy prior to enza and a repeat biopsy at the time of progression and whose tumor cells underwent RNA-sequencing after laser capture microdissection were identified. All progression biopsies were performed prior to discontinuing enza, enabling one to identify resistance mechanisms induced by continued enza treatment.


The study design is shown in FIG. 1A. Patient demographic information and prior treatments are shown in Table 2. Bone was the most common site for both pre-treatment and progression biopsies. Eighteen of 21 patients had the same tissue type biopsied at progression. In eight patients, the exact same lesion was biopsied at baseline and progression (FIG. 1B, Table 3). The median time on enza treatment was 226 days. PSA response at 12 weeks and the time between biopsies for each patient are shown in FIG. 1C.


To understand sample-to-sample differences, unsupervised hierarchical clustering was performed to find the nearest neighbor of 13/21 (62%) baseline samples and their matched progression sample pair (FIG. 2A). Samples did not cluster together based solely on the site of biopsy, indicating laser capture microdissection removed much of the microenvironment from these samples. Furthermore, whether the same lesion was biopsied did not impact how samples clustered.


Measurements of interest were examined in all the matched samples (FIG. 2B). To estimate AR transcriptional activity, Virtual Inference of Protein-activity by Enriched Regulon (VIPER) master regulator analysis was used 26. Nine (43%) patients did not have a marked difference in inferred AR activity. Nine (43%) patients had decreased AR activity, and three (14%) patients had increased AR activity at progression (FIG. 5A). A second method to measure AR activity—the ARG10 signature was used 27. ARG10 strongly correlated with the VIPER results (FIG. 5B). Though AR-V7 expression increased in several samples at progression, the difference in expression using the entire 21-patient cohort was not statistically significant (FIG. 5C).


Five clusters of CRPC tumors have been identified by RNA-sequencing analysis 25. Cluster 2 was enriched for tumors with loss of AR activity, increased E2F1 activity, and contained a preponderance of tumors that had lost AR expression 25, consistent with lineage plasticity. A subset of cluster 2 tumor samples was labeled NEPC based upon their morphologic appearance resembling small cell prostate cancer 25, though many of these tumor samples did not express canonical NEPC markers such as chromogranin A (CHGA) or synaptophysin (SYP) 25.


In examining the RNA-sequencing results from the baseline tumors, four of the five Aggarwal clusters were represented (clusters 1, 3, 4, and 5) in at least one sample, while no baseline sample harbored a cluster 2 program. The Labrecque transcription-based classifier that was developed on rapid autopsy CRPC samples was used to identify five subsets of prostate cancer: AR-driven prostate cancer (ARPC), amphicrine prostate cancer with neuroendocrine gene expression concomitant with AR signaling, AR-activity low prostate cancer, DNPC, and NEPC 24. The Labrecque classifier designated all the baseline samples in our cohort as ARPC.


To determine if any of the progression tumors in the cohort underwent lineage plasticity after enza, the Aggarwal cluster and Labrecque classifier designation were determined. Twelve of 21 matched pairs did not change their Aggarwal cluster designation. However, three of the 21 progression tumors (hereafter referred to as converters) had gene expression profiles consistent with cluster 2, supporting enza-induced conversion to an alternate lineage. The Labrecque classifier was also examined on the progression samples. The three converter samples designated as Aggarwal cluster 2 at progression were most consistent with DNPC by the Labrecque classifier, corroborating lineage plasticity in these tumors (FIG. 2B).


Additional gene signatures linked previously to lineage plasticity in progression vs. baseline biopsies were examined Comparing samples from the three converter patients, signature scores for genes upregulated in NEPC tumors described by Beltran, et al. 22 were increased (FIG. 5D). A previously described basal stemness signature 28 was also activated in these three progression samples (FIG. 5E). A 76 gene AR-repressed gene signature that was activated in a CRPC cell line that undergoes enza-induced lineage plasticity 29 was also increased in the progression samples from the three converters (FIG. 5F). Finally, predicted AR activity was significantly decreased in the progression samples from the converters by both VIPER and ARG10 signatures (FIG. 5A, 1G). In examining pre- and post-treatment samples using the entire 21-patient cohort, none of these signatures was significantly changed, demonstrating that activation of these lineage plasticity signatures was not a generalized effect of enza treatment. Altogether, these results suggest that enza-induced lineage plasticity and conversion to an AR-independent program occurs in a subset of tumors ( 3/21 or 14%), similar to the frequency of cluster 2 tumors (10%) described by Aggarwal previously25.


Notably, the baseline tumors from the three converter patients did not fall into the same Aggarwal cluster (cluster 4 for sample 80 and cluster 5 for samples 135, 210). The baseline tumors from these three patients did not cluster together using unsupervised clustering (FIG. 5H, 1I). These data indicate that there may be different starting points to lineage plasticity with enza treatment.


To identify genes linked with risk of lineage plasticity after enza, the differentially expressed genes between the three baseline samples from converters vs. the 18 non-converters were examined Pathway analysis implicated activation of MYC targets, E2F targets, and allograft rejection in baseline tumors from converters (FIG. 3A). There were no significantly downregulated pathways in baseline tumors from converters. To identify differentially activated transcription factors, master regulator analysis was performed. E2F1 was the top transcription factor predicted to be activated in the baseline tumors from converters (FIG. 3B, Table 4). Additionally, it was found that there was an upward trend in a previously described RB1 loss signature 31 in the progression samples from converters, further supporting that E2F1 activation contributes to the lineage switch (FIG. 5J). Other highly activated transcription factors in the baseline samples from converters include MYC family members and E2F4. Conversely, TP53—whose loss has been linked to lineage plasticity28, 32-33—was predicted to be the most deactivated transcription factor (FIG. 3B).


Next, genes that were significantly upregulated in the baseline tumors from converters vs. non-converters were identified. A 14-gene signature highly activated in the three baseline tumors from converters was identified (Table 5). Genes in this signature include those linked to: the Wnt pathway (RNF43 32 and TRABD2A 33), the spliceosome (SNRPF 34), and the electron transport chain (NDUFA12 35 and ATPSB 36). This signature trended downwards in the progression vs. baseline biopsies from the three converters (FIG. 6). These results indicate that this signature is not simply identifying tumor cells that have already undergone lineage plasticity prior to enza treatment. Rather, these genes may be markers of a transition state in cells susceptible to lineage plasticity.


Dividing the baseline samples between converters and non-converters, a cut off for this 14-gene lineage plasticity risk signature that separated the groups was defined (FIG. 3C). Additional cohorts with matched biopsies before and after enza with lineage plasticity information are lacking. However, it was hypothesized that patients whose baseline tumors had high scores for this lineage plasticity risk signature would have worse outcomes. Survival data from the time of ARSI treatment were available for several CRPC cohorts whose tumors had undergone RNA-sequencing—the International Dream Team dataset 10 and a prior prospective enza clinical trial led by our group 18. Because a subset of the patients in that latter enza clinical trial overlapped with the patients in this current report, patients from that clinical trial not represented here were analyzed. Using the pre-defined 14-gene signature score cut-off from the matched biopsy cohort, it was determined that high scores were associated with worse overall survival from the time of ARSI treatment in both independent datasets (p=0.076, p=0.006; FIG. 3D, E). Thus, high expression of the 14-gene lineage plasticity risk signature is linked to poor patient outcomes after ARSI treatment in CRPC. To determine if the lineage plasticity risk signature was activated in primary tumors, the TCGA dataset 39 was examined Importantly, only two of 495 patients had high risk scores (FIG. 6B). The lower frequency in primary tumors vs. CRPC cohorts suggests that activation of this lineage plasticity risk program may be induced by castration.


Validation datasets with matched biopsies before and after ARSI treatment that include information on lineage at time of progression are lacking. However, the impact of surgical castration on adenocarcinoma patient-derived xenografts (PDX) has been determined 23. Nine PDXs did not undergo castration-induced lineage plasticity, while one PDX—LTL331—does and converts to a resistant version called LTL331R 23. The patient from whom the LTL331 PDX is derived had evidence of lineage plasticity in his tumor when it became castration-resistant, demonstrating this model's fidelity 23,37. The lineage plasticity risk signature was highly activated in LTL331 vs. the other hormone naïve PDXs that do not undergo castration-induced lineage plasticity (FIG. 3F, 6C,D). Indeed, LTL331 was the only PDX whose lineage plasticity risk score was greater than the cut-off defined in the matched biopsy cohort (FIG. 3F). Prior work demonstrates that the exome of LTL331 is strikingly similar to its castration-induced lineage plasticity derivative, strongly suggesting that transdifferentiation—rather than clonal selection—may explain conversion in this tumor 23. Finally, the lineage plasticity risk score decreased in LTL331R vs. LTL331 (FIG. 6C), similar to the pattern observed in the progression vs. baseline samples from converters in our matched biopsy cohort (FIG. 6A).


Next, changes induced by enza between the baseline and progression samples from the three converters were investigated. The top differentially expressed genes are shown in FIG. 4A. The AR, AR target genes (KLK2, KLK3, and TMPRSS2), and the AR coactivator HOXB13 had markedly decreased expression (FIG. 4A, Table 6). In keeping with this, progression biopsies from converters had significantly reduced expression of AR target genes from the ARG10 gene signature 27 (FIG. 4B). Genes from the Beltran NEPC Upregulated signature were increased in progression samples from converters (FIG. 5B). It is worth noting that this signature contains both canonical NEPC genes and genes not explicitly associated with acquisition of neuroendocrine features that are AR-repressed. Specifically, examining canonical NEPC markers such as SYP, CHGA, and NCAM1, it was found that these genes were not highly upregulated at progression (Table 7). Other genes linked to NEPC (SYT11, CIITA, and ETVS) 22 or those normally repressed by the AR (CDCA7L, FRMD3, IKZF3, and TNFAIP2) 29 were more highly-expressed in the progression biopsies, indicating that these three converter tumors may be farther along the lineage plasticity spectrum than the previously described non-neuroendocrine DNPC subtype but not as far along as de novo NEPC or NEPC found at rapid autopsy by Labrecque, et al. 24 that harbor a more complete neuroendocrine program.


Pathway analysis between baseline and progression samples from the three converters demonstrated enrichment in several pathways, including: allograft rejection, interferon gamma response, interferon alpha response, and IL6/JAK/STAT signaling (FIG. 4C). Conversely, androgen and estrogen response—both linked to luminal differentiation—were the most downregulated, confirming loss of AR-dependence. Differences in gene expression between baseline and progression samples from the 18 patients whose tumors did not undergo lineage plasticity were examined. Several of the pathways activated in the converter tumors were also activated in the non-converters—namely, interferon alpha response, interferon gamma response, and TNF-α signaling (FIG. 9). Uniquely upregulated pathways in the converters include: allograft rejection, IL6-JAK-STAT3 signaling, inflammatory response, and complement. Uniquely downregulated pathways in the progression samples from non-converters included: E2F targets, G2M checkpoint, and hedgehog signaling. The only uniquely upregulated pathway in non-converters was protein secretion while uniquely downregulated pathways included hedgehog signaling, G2M checkpoint and E2F targets.


To understand the architecture of the tumors from the three converters, multiplex immunofluorescence (IF) was used with three luminal lineage markers (AR, NKX3.1, and HOXB13)—all downregulated at the mRNA level by RNA-sequencing (FIG. 4A)—and the NEPC marker INSM1 38. LuCaP PDX samples were used as positive and negative controls (FIG. 8B). Matched tissue samples for multiplex IF were available for subjects 135 and 210 but not for subject 80. One additional WCDT subject (103) with matched biopsies whose tumor underwent rapid clinical progression after enza treatment in the setting of a falling serum PSA—a clinical marker of AR-independence was identified. Matched RNA-sequencing was not available for this subject, but his tumor exhibited evidence of lineage plasticity (FIG. 4D). Representative staining images and quantitation of these markers are shown in FIG. 4D. There was a spectrum of AR, NKX3.1, and HOXB13 expression in baseline samples with some cells expressing low levels of each marker, while other cells expressed higher levels. However, at progression, there was a convergence towards population-wide loss of AR, NKX3.1, and HOXB13 in each sample. INSM1 upregulation was not identified in any of the baseline or progression tumors (FIG. 8C). These results match RNA-sequencing that failed to demonstrate upregulation of other canonical NEPC markers (Table 7) and that characterized the three converter samples as DNPC by the Labrecque classifier, rather than NEPC (FIG. 2B).


Finally, to determine if the progression samples from converters represented distinct clones with unique genetic alterations vs. baseline, DNA mutation and copy number analysis were performed. For subjects 80 and 103, the same tumor lesion was biopsied at baseline and progression. DNA-sequencing of these biopsies showed identical DNA mutations. For subjects 135 and 210, matched metastatic biopsy DNA-sequencing was unavailable. However, cell-free DNA was available. DNA-sequencing of cell-free DNA samples showed that mutations and copy number alterations were conserved between baseline and progression samples (Table 1).


Loss of the tumor suppressor genes TP53, RB1, and PTEN has been linked to lineage plasticity risk in pre-clinical models32, 33. However, it is not known if the presence of these genomic abnormalities in patient tumors is associated with risk of lineage plasticity to DNPC. One of the three converter patients (subject 80) was found to have an inactivating PTEN mutation and a second patient (subject 103) had RB1 loss, but none were found to have compound TP53/RB1/PTEN loss. When available, TP53/RB1/PTEN status for tumors from the Abida, et al.10 and Alumkal, et al.18 cohorts that had high lineage plasticity risk scores was examined. Of the seven high lineage plasticity risk score tumors examined from these two validation cohorts, only two tumors had loss of two or more of the genes TP53, RB1, and PTEN (Table 9). DNA-sequencing of matched metastatic biopsies for the cohort as a whole is shown in Table 10.









TABLE 1







DNA sequencing of matched samples from converters


demonstrates conserved alterations.









Patient ID
Mutation
Copy number gain/loss





DTB_80_BL
PTEN



DTB_80_Pro
PTEN


DTB_103_BL
RB1, FGFR3, NOTCH1


DTB_103_Pro
RB1, FGFR3, NOTCH1


DTB_135_BL
SPEN, FAT1
AR amplification, MYC




amplification


DTB_135_Pro
SPEN, FAT1, CTNNB1
AR amplification, MYC



(subclonal)
amplification


DTB_210_BL
APC, SPOP, KMT2C


DTB_210_Pro
APC, SPOP, KMT2C
















TABLE 2







Patient demographics and clinical information summary










Patients
n = 21















Median age at time of enrollment (SD)
71
(58-88)










Gleason score at diagnosis




≥8
16



 <8
5



ECOG performance status (%)



  0
10



  1
11



Metastatic site biopsied baseline (progression)











Bone
9
(9)



Lymph node
7
(8)



Pelvic soft tissue
2
(1)



Bladder wall
1
(0)



Liver
1
(2)



Adrenal
1
(1)










Same lesion biopsied
8



Visceral metastatic disease at time of biopsy
5



Prior treatment



Abiraterone
7



ADT
21



Bicalutamide
8



Cabazitaxel
1



Docetaxel
3



Sip-T
1











Median PSA at enrollment (SD)
57
(200)










50% PSA response to enzalutamide
7











Median time on enzalutamide, days (SD)
261
(315)

















TABLE 3







Patient and biopsy information















Baseline


Time

PSA




biopsy
Progression
Same site
Between
PSA at
Change at
Prior


Sample_ID
tissue
tissue
biopsied
Biopsies
baseline
12 weeks
treatment

















DTB_022
Bone
Bone
No
85
7.28
N/A
ADT,









abiraterone


DTB_024
Liver
Liver
No
99
48.92
75.05
ADT,









abiraterone,









docetaxel


DTB_060
Adrenal
Adrenal
Yes
449
102.45
−20.41
ADT


DTB_063
LN
LN
No
368
20.9
−74.64
ADT


DTB_073
Bone
Bone
No
54
57.67
156.81
ADT,









Abiraterone


DTB_080
LN
LN
Yes
266
14.92
−28.48
ADT


DTB_089
Bone
Liver
No
91
14.27
181.18
ADT,









Abiraterone


DTB_098
LN
LN
Yes
615
148.39
−83.74
ADT


DTB_102
Bladder
LN
No
533
1210.48
−93.02
ADT,









abiraterone,









docetaxel,









cabazitaxel


DTB_111
LN
LN
Yes
134
35.02
113.99
ADT,









Abiraterone


DTB_127
LN
LN
Yes
226
228
169.29
ADT,









Abiraterone


DTB_135
LN
LN
No
73
9.19
164.26
ADT,









bicalutamide


DTB_137
Bone
Bone
No
441
539.62
−10.85
ADT,









bicalutamide


DTB_141
Bone
Bone
No
285
140.07
−81.04
ADT,









bicalutamide


DTB_149
Bone
Bone
No
262
16.45
−80.29
ADT,









bicalutamide


DTB_167
Soft
Bone
No
827
136.64
−88.38
ADT,



tissue





bicalutamide,









sipuleucel-T


DTB_176
Soft
Soft
Yes
291
3.57
−64.76
ADT,



tissue
tissue




bicalutamide


DTB_194
Bone
Bone
No
88
103.55
100.36
ADT,









bicalutamide


DTB_210
Bone
Bone
No
200
70.45
−83.36
ADT


DTB_232
Bone
Bone
Yes
114
5.31
−0.55
ADT,









docetaxel


DTB_265
LN
LN
Yes
105
42.93
5.84
ADT,









bicalutamide
















TABLE 4







Converter vs. non-converter baseline Master Regulator analysis












Regulon
Size
NES
p.value















ABL1
ABL1
29
0.051236
0.959138


ACTB
ACTB
22
−1.01068
0.312171


AHR
AHR
195
−1.74628
0.080762


AIF1L
AIF1L
30
−0.03049
0.97568


ANG
ANG
16
−0.20733
0.83575


APOB
APOB
13
0.23179
0.816701


AR
AR
504
−2.00987
0.044445


ARID3A
ARID3A
15
1.15268
0.249042


ARNT
ARNT
20
1.271309
0.203619


ARVCF
ARVCF
24
−0.92051
0.357304


ASCL1
ASCL1
25
−0.90166
0.367238


ATF1
ATF1
60
−0.67157
0.501858


ATF2
ATF2
111
−2.28452
0.022341


ATF3
ATF3
256
2.636052
0.008388


ATF4
ATF4
77
1.324082
0.185476


ATF6
ATF6
48
0.596372
0.550927


ATOH1
ATOH1
11
−0.79886
0.42437


BACH1
BACH1
19
−0.64787
0.517068


BARX2
BARX2
16
−1.15849
0.246664


BATF
BATF
117
1.915968
0.055369


BCL11A
BCL11A
65
−0.45344
0.650232


BCL3
BCL3
187
2.151217
0.031459


BCL6
BCL6
77
−0.44056
0.659529


BCLAF1
BCLAF1
139
2.42129
0.015466


BCR
BCR
47
−0.28628
0.774662


BDP1
BDP1
68
−0.69727
0.485632


BHLHE40
BHLHE40
33
−1.88141
0.059916


BMP2
BMP2
269
−2.994
0.002753


BRCA1
BRCA1
168
2.87972
0.00398


BRF1
BRF1
25
−0.26372
0.791995


BRF2
BRF2
30
−0.24797
0.804161


CBX5
CBX5
16
−0.13761
0.890549


CCDC116
CCDC116
47
0.152711
0.878626


CCL20
CCL20
15
0.481348
0.630269


CCNT2
CCNT2
78
0.109371
0.912908


CDC45
CDC45
18
−0.74948
0.453568


CEBPB
CEBPB
429
2.898243
0.003753


CEBPZ
CEBPZ
73
−0.56453
0.572395


CHD2
CHD2
210
2.415137
0.015729


CIC
CIC
14
−1.52926
0.1262


CLIC1
CLIC1
20
−0.44992
0.652767


CLOCK
CLOCK
29
1.260735
0.207404


COMT
COMT
19
−0.71566
0.474203


CREB1
CREB1
127
−0.53051
0.595761


CREM
CREM
48
0.069052
0.944948


CRKL
CRKL
36
−0.32866
0.742414


CSNK2B
CSNK2B
15
−1.19408
0.232447


CTBP2
CTBP2
110
−2.37744
0.017433


CTCF
CTCF
1030
3.206967
0.001341


CUX1
CUX1
405
3.125283
0.001776


DACH1
DACH1
152
2.282266
0.022474


DBP
DBP
62
1.594974
0.110718


DCAF11
DCAF11
21
−0.62929
0.529162


DDAH2
DDAH2
15
0.361102
0.718024


DDIT3
DDIT3
134
0.499877
0.617162


DGCR8
DGCR8
26
−0.23777
0.812056


DHRS2
DHRS2
27
−0.52634
0.598652


DLX2
DLX2
36
−1.9245
0.054292


DLX5
DLX5
23
−0.33429
0.738162


E2F1
E2F1
740
8.39697
4.58E−17


E2F2
E2F2
24
1.350676
0.176799


E2F3
E2F3
33
−0.65908
0.509844


E2F4
E2F4
260
6.889355
5.60E−12


E2F6
E2F6
260
3.302534
0.000958


EBF1
EBF1
184
−0.85838
0.390681


EEF1A1
EEF1A1
15
−1.15332
0.248781


EGR1
EGR1
540
−1.50333
0.132754


EGR2
EGR2
58
−0.64473
0.519102


EGR3
EGR3
11
−0.43829
0.661177


ELF1
ELF1
379
−1.2287
0.219183


ELF3
ELF3
36
0.866801
0.386051


ELK1
ELK1
177
1.107087
0.268256


ELK3
ELK3
23
−1.03345
0.301394


ELK4
ELK4
93
−0.6164
0.537629


EN1
EN1
16
−0.931
0.351853


EP300
EP300
427
−1.49352
0.1353


EPAS1
EPAS1
71
−0.74583
0.455769


ERF
ERF
23
−0.18083
0.856505


ERG
ERG
45
−0.61526
0.538381


ESR1
ESR1
389
−2.25367
0.024217


ESR2
ESR2
90
−0.89841
0.36897


ESRRA
ESRRA
69
0.088802
0.929239


ETS1
ETS1
538
2.444806
0.014493


ETS2
ETS2
60
−1.10991
0.267037


ETV4
ETV4
33
0.985929
0.324168


ETV6
ETV6
61
0.012768
0.989813


EVX1
EVX1
11
−1.35387
0.175777


EZH2
EZH2
54
1.776698
0.075618


FAM78A
FAM78A
17
−0.16198
0.871318


FANK1
FANK1
31
−0.1858
0.852601


FBXO31
FBXO31
17
−1.09211
0.274787


FIBCD1
FIBCD1
20
−0.36686
0.713722


FLI1
FLI1
64
−2.14641
0.03184


FOS
FOS
574
−1.79691
0.072349


FOSB
FOSB
24
0.004458
0.996443


FOSL1
FOSL1
120
0.130066
0.896514


FOSL2
FOSL2
85
1.365824
0.171994


FOXA1
FOXA1
383
−1.09373
0.274075


FOXA2
FOXA2
213
1.892128
0.058474


FOXC1
FOXC1
29
−1.81002
0.070292


FOXC2
FOXC2
32
−0.90415
0.365917


FOXL2
FOXL2
21
−1.18545
0.235839


FOXM1
FOXM1
82
2.675307
0.007466


FOXN1
FOXN1
33
−0.97553
0.329296


FOXO1
FOXO1
142
−2.32375
0.020139


FOXO3
FOXO3
106
−1.0564
0.290786


FOXO4
FOXO4
26
0.560582
0.575083


FOXP3
FOXP3
85
0.971092
0.331502


GABPA
GABPA
366
2.163614
0.030494


GATA1
GATA1
280
0.462909
0.64343


GATA2
GATA2
506
1.542317
0.122997


GATA3
GATA3
511
−0.55035
0.582077


GATA4
GATA4
70
−1.49867
0.133958


GATA6
GATA6
40
−0.45295
0.650588


GFI1
GFI1
21
1.522495
0.127885


GLI1
GLI1
109
−1.6911
0.090817


GLI2
GLI2
73
−0.93476
0.34991


GLI3
GLI3
48
−1.20849
0.22686


GMPR2
GMPR2
14
0.516862
0.605253


GNAZ
GNAZ
16
−0.51571
0.606054


GNB1L
GNB1L
16
−1.46036
0.144191


GP1BB
GP1BB
22
−0.18902
0.850079


GTF2B
GTF2B
231
3.288553
0.001007


GTF2F1
GTF2F1
76
2.911749
0.003594


HBP1
HBP1
32
0.666748
0.504933


HDAC2
HDAC2
89
0.702261
0.482516


HES1
HES1
70
0.4195
0.674851


HESX1
HESX1
11
0.48202
0.629792


HEY1
HEY1
18
−0.88366
0.37688


HHEX
HHEX
14
−0.20373
0.838562


HIF1A
HIF1A
261
−1.22063
0.222228


HIF3A
HIF3A
11
−0.61827
0.5364


HIRA
HIRA
19
0.89105
0.372902


HIST1H2AB
HIST1H2AB
16
0.839262
0.401322


HIST1H2AD
HIST1H2AD
18
0.032375
0.974173


HIST1H2AG
HIST1H2AG
18
−0.5441
0.586373


HIST1H2AH
HIST1H2AH
14
−0.99929
0.317654


HIST1H2BD
HIST1H2BD
17
−1.04682
0.295181


HIST1H2BF
HIST1H2BF
17
−0.43296
0.665044


HIST1H2BJ
HIST1H2BJ
18
−0.86077
0.389362


HIST1H3B
HIST1H3B
16
−0.37424
0.708223


HIST1H3D
HIST1H3D
18
−0.26053
0.794458


HIST1H4B
HIST1H4B
17
0.253817
0.799637


HIST1H4I
HIST1H4I
19
0.385611
0.699785


HLX
HLX
14
−0.95844
0.337843


HMGA1
HMGA1
52
0.141333
0.887607


HMGN3
HMGN3
55
0.779959
0.435415


HNF1A
HNF1A
78
−1.74654
0.080718


HNF1B
HNF1B
48
0.274363
0.783806


HNF4G
HNF4G
102
0.268923
0.787989


HOXA10
HOXA10
29
−0.47125
0.637461


HOXA11
HOXA11
14
−1.41179
0.158011


HOXA5
HOXA5
20
−2.78743
0.005313


HOXA9
HOXA9
38
1.060151
0.289076


HOXC13
HOXC13
15
−0.82797
0.40769


HOXC8
HOXC8
15
−1.58404
0.113184


HOXD13
HOXD13
23
2.051993
0.04017


HSF1
HSF1
115
0.134176
0.893263


HSPA1B
HSPA1B
18
−0.63053
0.528346


ID1
ID1
142
−2.86808
0.00413


ID2
ID2
71
−2.23042
0.025719


ID3
ID3
63
−2.84359
0.004461


IFI16
IFI16
17
−0.04101
0.967291


IRF1
IRF1
311
1.058916
0.289638


IRF2
IRF2
34
−0.08632
0.931208


IRF3
IRF3
179
1.663856
0.096141


IRF4
IRF4
53
1.16537
0.243869


IRF5
IRF5
27
−0.08125
0.935243


IRF6
IRF6
1203
1.284136
0.199095


IRF7
IRF7
33
−0.23835
0.811613


IRF8
IRF8
47
0.528977
0.596822


ISL1
ISL1
13
−0.43434
0.66404


JUN
JUN
673
3.060137
0.002212


JUNB
JUNB
115
−0.81085
0.417451


JUND
JUND
359
3.282218
0.00103


KAT2A
KAT2A
35
1.340154
0.180195


KLF1
KLF1
20
−1.61554
0.106194


KLF10
KLF10
32
−1.68154
0.092658


KLF15
KLF15
13
0.50017
0.616956


KLF2
KLF2
39
−0.88487
0.376225


KLF4
KLF4
126
−1.3626
0.173009


KLF5
KLF5
38
1.33999
0.180249


KLF6
KLF6
35
−1.87978
0.060138


KLF9
KLF9
11
−2.04365
0.040988


LEF1
LEF1
65
−0.32435
0.745672


LHX2
LHX2
26
0.21167
0.832364


MAF
MAF
36
0.15297
0.878422


MAFF
MAFF
119
0.079839
0.936365


MAFK
MAFK
177
0.184701
0.853464


MAPK1
MAPK1
26
−1.38973
0.16461


MAX
MAX
413
6.973296
3.10E−12


MAZ
MAZ
12
0.52102
0.602353


MEF2A
MEF2A
69
−0.3792
0.704537


MEF2C
MEF2C
52
1.242668
0.21399


MEF2D
MEF2D
13
−0.36941
0.71182


MEIS1
MEIS1
24
−0.62118
0.534482


MEIS2
MEIS2
20
−1.95293
0.050828


MITF
MITF
80
−0.92177
0.356649


MLXIPL
MLXIPL
9
0.399449
0.689563


MRPL40
MRPL40
18
−0.63824
0.523315


MSC
MSC
87
−0.49271
0.622218


MSX1
MSX1
25
−2.40653
0.016105


MSX2
MSX2
55
−1.27053
0.203895


MTF1
MTF1
27
−0.33412
0.738291


MTRNR2L1
MTRNR2L1
16
−0.61432
0.539004


MXD1
MXD1
21
0.771096
0.44065


MXI1
MXI1
68
1.548788
0.121433


MYB
MYB
89
2.119714
0.03403


MYBL2
MYBL2
49
1.329846
0.183569


MYC
MYC
1307
6.630539
3.34E−11


MYCN
MYCN
107
−0.92136
0.356862


NANOG
NANOG
186
−2.41909
0.015559


NBPF1
NBPF1
30
−1.38118
0.167224


NCOA1
NCOA1
15
−0.91836
0.358429


NCOA3
NCOA3
46
−0.54863
0.583259


NFAT5
NFAT5
34
0.110771
0.911798


NFATC1
NFATC1
51
−0.00658
0.994749


NFATC4
NFATC4
22
−0.47439
0.635224


NFE2
NFE2
133
0.230329
0.817836


NFIC
NFIC
24
−1.22856
0.219239


NFIX
NFIX
18
−0.30978
0.756726


NFKB1
NFKB1
126
0.032244
0.974278


NFYA
NFYA
258
2.454322
0.014115


NFYB
NFYB
285
0.388634
0.697547


NKRF
NKRF
11
−0.8646
0.38726


NKX2-1
NKX2-1
20
0.4038
0.68636


NR1I2
NR1I2
14
1.005625
0.314596


NR1I3
NR1I3
51
−1.25017
0.211239


NR2C2
NR2C2
214
2.498588
0.012469


NR2F1
NR2F1
25
−0.93504
0.34977


NR2F2
NR2F2
42
−1.22894
0.219096


NR3C1
NR3C1
275
−0.67159
0.501843


NR3C2
NR3C2
40
−0.55382
0.579703


NR4A1
NR4A1
541
−3.11662
0.001829


NR4A2
NR4A2
45
−1.72408
0.084694


NR5A2
NR5A2
21
−1.49924
0.133812


NR6A1
NR6A1
9
−0.69796
0.485204


NRF1
NRF1
404
4.353719
1.34E−05


NUP214
NUP214
23
−0.51712
0.605073


PAWR
PAWR
22
−0.1726
0.862968


PAX2
PAX2
37
−0.19594
0.844658


PAX5
PAX5
159
0.74832
0.454267


PAX6
PAX6
81
2.569159
0.010195


PAX8
PAX8
42
−1.19595
0.231717


PBX1
PBX1
43
−1.56217
0.118249


PBX3
PBX3
257
−0.15059
0.880296


PDE4DIP
PDE4DIP
30
−0.84827
0.396287


PDX1
PDX1
35
−3.06466
0.002179


PGR
PGR
74
−1.70675
0.087868


PI4KA
PI4KA
21
−0.49943
0.617479


PITX1
PITX1
15
−1.05999
0.289151


PITX2
PITX2
82
−1.12354
0.261207


PLEK
PLEK
15
−0.70927
0.47816


PMF1
PMF1
24
−1.44535
0.148359


POLR3A
POLR3A
19
0.922368
0.356337


POU1F1
POU1F1
20
0.123814
0.901463


POU2F1
POU2F1
45
2.369076
0.017833


POU2F2
POU2F2
119
1.59509
0.110692


POU3F2
POU3F2
168
−3.55876
0.000373


POU5F1
POU5F1
129
−1.57145
0.116079


POU6F1
POU6F1
11
0.059949
0.952197


PPARA
PPARA
277
−1.05335
0.29218


PPARD
PPARD
111
−0.94907
0.342584


PPARG
PPARG
304
−1.84617
0.064867


PPARGC1A
PPARGC1A
29
−0.56708
0.570663


PPIL2
PPIL2
20
−0.71742
0.473113


PRAME
PRAME
22
0.271773
0.785796


PRDM1
PRDM1
43
−1.98139
0.047547


PROX1
PROX1
29
0.171039
0.864193


RAB36
RAB36
17
−0.57724
0.563775


RAD21
RAD21
473
0.404801
0.685624


RARA
RARA
61
−1.44521
0.1484


RARB
RARB
44
−3.47311
0.000514


RARG
RARG
27
−0.24677
0.805085


RBPJ
RBPJ
70
−0.82953
0.406805


REL
REL
87
0.886843
0.375164


RELA
RELA
106
−1.10166
0.270611


RELB
RELB
51
−0.68492
0.493391


REST
REST
172
−0.67626
0.498874


RFX1
RFX1
14
−0.57575
0.564781


RFX5
RFX5
84
1.307749
0.190959


RORA
RORA
19
0.781685
0.434399


RUNX1
RUNX1
245
−0.22748
0.82005


RUNX2
RUNX2
151
−1.82508
0.067989


RUNX3
RUNX3
65
0.131457
0.895413


RXRA
RXRA
72
1.280295
0.200442


SALL1
SALL1
14
−1.18
0.238001


SATB1
SATB1
10
0.462061
0.644038


SDF2L1
SDF2L1
23
−0.87616
0.380941


SETBP1
SETBP1
59
−0.14267
0.886549


SETDB1
SETDB1
125
−0.46296
0.643395


SIM2
SIM2
36
0.467962
0.639811


SIN3A
SIN3A
55
2.605818
0.009166


SIRT6
SIRT6
39
1.743563
0.081235


SIX1
SIX1
18
1.231365
0.218186


SIX5
SIX5
263
−0.06625
0.947176


SKI
SKI
24
−0.59419
0.552385


SMAD1
SMAD1
75
−1.54514
0.122314


SMAD2
SMAD2
431
−3.11924
0.001813


SMAD3
SMAD3
510
−3.35926
0.000782


SMAD4
SMAD4
167
−2.0752
0.037968


SMAD5
SMAD5
56
−1.43085
0.152472


SMARCA4
SMARCA4
125
4.156509
3.23E−05


SMARCB1
SMARCB1
117
2.335056
0.01954


SMC3
SMC3
75
1.168808
0.242481


SNAI2
SNAI2
64
−2.32188
0.02024


SOX13
SOX13
12
−0.26968
0.787409


SOX17
SOX17
36
−1.07345
0.283068


SOX2
SOX2
184
−1.84782
0.064629


SOX4
SOX4
26
−0.22655
0.820776


SOX9
SOX9
174
−1.80944
0.070383


SP1
SP1
865
−2.47968
0.01315


SP100
SP100
24
−0.61601
0.537885


SP2
SP2
188
1.211764
0.225603


SP3
SP3
150
1.6031
0.108913


SP4
SP4
29
−1.69027
0.090977


SPI1
SPI1
284
−0.15241
0.878865


SREBF1
SREBF1
170
1.139298
0.254579


SREBF2
SREBF2
78
0.503728
0.614453


SRF
SRF
293
1.940434
0.052327


STAT1
STAT1
383
0.842839
0.399319


STAT2
STAT2
88
0.283629
0.776695


STAT3
STAT3
598
1.421507
0.155169


STAT4
STAT4
21
1.011493
0.311781


STAT5A
STAT5A
168
1.104389
0.269425


STAT5B
STAT5B
25
−1.52632
0.12693


STAT6
STAT6
77
−0.23499
0.814219


SUZ12
SUZ12
151
−0.80984
0.418032


TAF1
TAF1
439
5.82917
5.57E−09


TAF7
TAF7
70
−1.03045
0.302799


TAL1
TAL1
162
−1.26458
0.206021


TBP
TBP
184
4.1589
3.20E−05


TBX2
TBX2
39
0.630216
0.528553


TBX3
TBX3
25
0.495094
0.620534


TBX5
TBX5
18
−2.56321
0.010371


TCF12
TCF12
165
−0.39208
0.694998


TCF3
TCF3
12
−1.11343
0.265524


TCF4
TCF4
391
0.753801
0.450969


TCF7L1
TCF7L1
15
0.30834
0.757823


TCF7L2
TCF7L2
49
−0.91532
0.360021


TEF
TEF
23
−0.62575
0.531482


TFAM
TFAM
16
−0.27248
0.785252


TFAP2A
TFAP2A
385
1.33025
0.183436


TFAP2C
TFAP2C
153
2.376242
0.01749


TFE3
TFE3
18
−0.41694
0.67672


TGIF1
TGIF1
19
0.511829
0.608771


THAP1
THAP1
44
0.007277
0.994194


THRB
THRB
14
−0.46604
0.641183


TP53
TP53
1576
−6.57724
4.79E−11


TRIM28
TRIM28
45
0.695178
0.486944


TSC22D3
TSC22D3
61
−1.27922
0.200819


TWIST1
TWIST1
80
1.198189
0.230844


USF1
USF1
30
0.616252
0.537728


USF2
USF2
236
1.231038
0.218309


VDR
VDR
113
−0.72764
0.466833


XBP1
XBP1
66
0.414696
0.678365


XRCC4
XRCC4
28
−0.79178
0.428489


YBX1
YBX1
42
−0.63687
0.524206


YY1
YY1
410
3.781417
0.000156


ZBTB17
ZBTB17
20
−0.38443
0.700662


ZBTB33
ZBTB33
151
−1.95137
0.051013


ZBTB7A
ZBTB7A
163
0.938493
0.347991


ZEB1
ZEB1
118
−0.74247
0.457803


ZEB2
ZEB2
20
−0.89984
0.368205


ZNF143
ZNF143
30
−0.98471
0.324765


ZNF263
ZNF263
245
−0.16919
0.865649


ZNF274
ZNF274
65
0.593574
0.552797


ZZZ3
ZZZ3
21
0.038971
0.968913
















TABLE 5





14 Gene Lineage Plasticity Risk Signature

















RNF43



SNRPF



TRABD2A



NDUFA12



GAS2L3



RPS24



DNA2



RP5-857K21.10



POC1B



ADK



ATP5B



XPOT



SLCO1B3



RHOBTB1

















TABLE 6







Genes downregulated in progression versus baseline in converters












log2 fold




Gene
change
p adjusted















GJB1
−13.0109
0.00803528



KRT19
−12.4296
0.009995189



MSMB
−10.9755
1.60052E−12



KLK4
−10.8022
6.07743E−28



RFX6
−10.7898
2.69827E−09



SPDEF
−10.5975
1.95528E−06



PRAC1
−10.586
2.19387E−16



TRPM8
−10.432
6.84759E−10



CWH43
−10.4251
1.21947E−12



SFTPA2
−10.4246
6.36253E−14



KLK2
−10.12
 4.5295E−14



RP11-64K7.1
−9.84876
6.00493E−08



C1orf116
−9.5854
3.74956E−32



TRPV6
−9.46577
1.60052E−12



PCAT14
−9.45153
1.05491E−08



KLK3
−9.31504
5.91494E−38



LMAN1L
−9.2235
3.82153E−05



FAM155B
−9.12385
3.15397E−05



SFT2D3
−9.05073
0.014343495



CLDN8
−9.0205
0.000247776



PLPPR1
−9.01465
7.62753E−06



AR
−9.00786
2.67485E−20



CH17-335B8.6
−8.98895
 5.5873E−05



GCG
−8.9036
0.002364738



RP11-250B16.1
−8.8774
6.18409E−08



LUZP2
−8.84452
8.01584E−08



ELF5
−8.828
1.54204E−05



HOXB13
−8.64854
1.40708E−17



NKX3-1
−8.6224
3.94512E−21



RP11-167H9.6
−8.57309
0.001755056



AP1M2
−8.56433
0.000299706



RP11-386M24.6
−8.49415
6.31748E−09



COLCA1
−8.4758
2.22009E−11



NUDT11
−8.42477
0.000117825



MAL2
−8.36678
 4.5655E−06



MAGEA1
−8.3178
0.025761966



ALDH3B2
−8.29458
0.000408765



ELFN2
−8.25871
 2.0192E−07



BMPR1B
−8.22216
1.13195E−09



SLC9A2
−8.22212
3.93176E−05



GDF15
−8.20863
3.24736E−08



RIPK4
−8.16098
0.000679994



RP6-201G10.2
−8.13008
0.009363237



MB
−8.08018
5.57196E−05



RP11-810K23.10
−8.06303
0.013754538



RP11-414J4.2
−8.04941
1.95528E−06



PRR36
−8.04044
0.002062771



GLYATL1
−8.03801
8.01584E−08



OR51E2
−8.03443
5.29546E−08



VSTM2L
−8.01538
0.001159402



RP11-191G24.2
−7.98022
0.000394074



CPNE4
−7.97461
1.81751E−05



ZG16B
−7.91661
8.89766E−06



STEAP2
−7.90516
6.27827E−19



TGM3
−7.90155
0.000201653



KB-1562D12.1
−7.85336
0.006725243



MUM1L1
−7.81099
0.047267105



FOXA1
−7.78551
 3.8039E−07



SSTR1
−7.75964
2.15594E−05



FOLH1
−7.73365
 2.8238E−05



NPY2R
−7.70357
0.001820183



GPR81
−7.67445
0.003315652



PLA2G4F
−7.6702
2.86056E−05



AP001615.9
−7.64992
0.00281238



RP11-664D7.4
−7.63551
2.30599E−06



TMPRSS2
−7.61592
1.57797E−12



CBLC
−7.5637
0.011752953



LONRF2
−7.55205
 7.8229E−08



PRAC2
−7.50435
0.006941479



KLKP1
−7.40505
2.49362E−05



CTD-2008P7.9
−7.40308
0.031968996



ZDHHC8P
−7.39731
0.000105089



PTPRT
−7.39084
1.53606E−08



KLK15
−7.38721
0.00151056



HOXA11-AS1
−7.38246
0.000318881



MSI1
−7.38125
0.009341293



RGS11
−7.35431
1.15968E−05



ITIH6
−7.30192
0.013754538



CNNM1
−7.295
 1.0376E−12



RP11-96O20.1
−7.28528
2.69827E−09



ESRP1
−7.27842
 3.9422E−06



RP11-44F14.8
−7.2738
0.000169539



OPRK1
−7.26447
0.011482303



OVOL1
−7.22031
0.019243098



RP11-23F23.2
−7.206
0.000510792



CTC-429C10.4
−7.19953
0.002543297



TSPAN1
−7.16544
1.77922E−12



B4GALNT4
−7.15865
0.015346028



HNF1B
−7.14867
0.006147776



DPY19L2P4
−7.14349
0.018382137



RP11-217E22.2
−7.09768
0.007346357



BRINP3
−7.09246
0.007726148



KCNQ4
−7.08721
0.01062877



LPAR3
−7.0795
2.32188E−09



RP11-429J17.8
−7.03507
 9.4023E−06



ACPP
−7.03063
2.29033E−08



NKAIN1
−7.01909
0.000140179



SLC6A11
−6.99287
1.19755E−05



CKMT1B
−6.97352
2.85313E−05



AC005077.14
−6.92458
0.012413722



EPCAM
−6.91034
1.49312E−05



CHMP4C
−6.90771
0.000103013



CLDN3
−6.88946
5.80941E−05



C8orf34
−6.87524
0.003315652



EHF
−6.86723
2.49362E−05



CLDN4
−6.8506
0.000421405



TMSB15A
−6.83338
0.022191671



SIM2
−6.81808
 3.0951E−05



CDH7
−6.80255
0.001317177



CREB3L1
−6.78654
5.38642E−19



RET
−6.76795
0.006752055



HS6ST3
−6.74427
0.000248805



SHANK2
−6.74014
2.30599E−06



TMEFF2
−6.73602
0.000219103



HSD17B6
−6.72389
3.63729E−05



RP11-810K23.9
−6.69377
0.048963223



RP11-386M24.3
−6.67882
0.011406542



IL20RA
−6.6682
0.000394074



CHRNA2
−6.66762
0.02380943



CRABP2
−6.64933
0.000520944



ZBED9
−6.59905
0.00365828



TSPAN8
−6.59719
0.01310687



DNASE2B
−6.5658
0.020111479



ARFGEF3
−6.5653
1.43252E−16



CTD-2315M5.2
−6.5475
0.001864013



CRISP3
−6.52223
0.007176935



PDZK1IP1
−6.51569
0.01055954



STEAP1
−6.49332
1.45863E−09



PBOV1
−6.48531
0.000131516



SPTBN2
−6.4581
0.000851134



RAB3B
−6.37894
1.62558E−08



SYT7
−6.37818
2.77749E−05



RP11-572M18.1
−6.3717
0.029303804



SEMA3C
−6.36548
6.49127E−19



COL2A1
−6.35909
 5.8515E−05



FRMPD4
−6.35095
0.046924645



RP4-568C11.4
−6.33688
0.000190283



OVOL2
−6.3276
0.042698538



CUX2
−6.25239
0.000319499



TFAP2C
−6.24995
0.007446394



RIPPLY3
−6.23858
0.025465136



DNAJC22
−6.15536
0.038581579



HIST3H2A
−6.14943
2.47998E−05



FAM3B
−6.12534
0.000166735



CHRM1
−6.10673
0.024066227



C1orf210
−6.10671
0.009161256



TRGC1
−6.10126
0.000368444



GRHL2
−6.07235
4.31758E−06



EPN3
−6.05967
0.004856004



CTD-2626G11.2
−6.03012
0.015236703



MAPK8IP2
−6.01836
0.002676092



CAMSAP3
−6.0032
0.018436013



GLB1L2
−5.98958
0.002169017



ARL4P
−5.98621
0.019243098



PKNOX2
−5.9705
0.001537648



PROM2
−5.95201
0.000150439



RP11-794G24.1
−5.93328
0.006307743



ARHGAP6
−5.90103
3.23698E−06



C3orf80
−5.88771
0.011706175



CERS1
−5.85838
0.007844838



KAZALD1
−5.85645
0.000413558



KCNG1
−5.84824
0.031523396



AGTR1
−5.83595
9.15929E−07



GAL
−5.83458
0.004055001



PPP1R1B
−5.82851
3.13653E−09



RANBP3L
−5.81794
2.51666E−06



PRSS8
−5.81696
0.005362879



PLPP1
−5.75925
2.67485E−20



GLYATL1P1
−5.75464
0.013457395



SAMD5
−5.74866
2.42795E−08



EPHA7
−5.72406
0.026780088



TACSTD2
−5.6688
1.03652E−05



PRR15L
−5.65178
0.001562489



F12
−5.63147
0.001590333



PYCR1
−5.59224
0.002212432



KRT8
−5.58961
0.001317177



KDF1
−5.58481
0.025166394



KLF15
−5.57358
0.000510792



RP1-239B22.5
−5.57151
0.030116743



XDH
−5.54559
0.000930733



ANKRD30A
−5.54261
0.001549402



KCNC2
−5.53619
0.035899302



SLC44A4
−5.53579
0.000389098



TMC4
−5.51246
0.001097835



CLDN1
−5.50523
0.00230044



NWD1
−5.47963
1.12618E−05



LAMA1
−5.47147
0.012383175



RP11-44F14.2
−5.47147
0.000254976



ERBB3
−5.45723
8.17457E−06



CAPN13
−5.44675
0.007996556



SH2D4A
−5.44673
0.00031238



GATA2
−5.43639
6.05752E−08



CRYM
−5.4186
0.003834794



HPN
−5.39975
0.000610051



ARHGEF38
−5.3921
 1.4071E−05



KLK11
−5.38859
0.042443384



TMEM125
−5.38713
0.008470304



RP11-61N20.3
−5.37734
0.020600986



RP11-887P2.1
−5.36726
0.00271796



ST6GALNAC1
−5.34964
0.002310415



BCYRN1
−5.34173
0.000394074



RORB
−5.33749
0.001313938



RP11-680C21.1
−5.33702
0.027161703



MUC13
−5.33548
0.008421801



UNC5A
−5.33402
0.014974489



WNT7B
−5.33287
0.028295215



MAOA
−5.3151
2.32964E−05



BRSK2
−5.31409
0.041483081



ONECUT2
−5.31276
0.002495169



PRR16
−5.29134
3.63871E−09



LAD1
−5.28453
0.00175777



TTC6
−5.28105
0.000764496



TRGV9
−5.274
0.001319505



ERVMER34-1
−5.2629
0.006941479



PDE9A
−5.25653
0.000256129



NFIX
−5.24719
 9.0469E−16



SLC45A3
−5.23953
6.09221E−05



KRT18
−5.23594
0.000916193



CGREF1
−5.2326
0.000520944



FAXC
−5.22737
0.004551851



RIMS1
−5.21989
0.023229561



CKMT1A
−5.21458
0.000510792



KIF5C
−5.21069
6.98142E−06



TUFT1
−5.19053
0.010580032



CGN
−5.18038
0.005195388



DCDC2
−5.16915
0.00601495



LYPD6B
−5.14802
0.03778331



SCNN1A
−5.13995
0.015531057



FRAS1
−5.13625
0.018371812



KCND3
−5.11214
2.50126E−06



AQP3
−5.08797
0.00041265



TBX3
−5.07588
0.000807785



PCDHB2
−5.06229
0.005053276



PLA2G2A
−5.0594
0.004403906



SLC30A4
−5.05773
2.11383E−07



DNAJC12
−5.05297
0.008854581



GSTO2
−5.03542
0.019916132



PCDHB16
−5.03101
0.001506409



MYH14
−5.00681
0.038148283



FAM47E-STBD1
−5.00384
0.001530784



OR7E47P
−4.99846
0.027205581



CGNL1
−4.99223
1.08457E−06



SV2C
−4.98913
0.00492018



RAMP1
−4.96574
0.019674785



ESRP2
−4.96328
0.000637971



ASIC1
−4.91851
8.98808E−05



LRRC26
−4.91831
0.025465136



RP11-159H10.3
−4.89845
0.022784159



01-Mar
−4.87761
1.33169E−05



C1orf168
−4.87428
0.010816456



ADGRV1
−4.86761
0.000834175



RP11-123K3.4
−4.84725
0.017085905



C9orf152
−4.84208
1.34448E−06



RAB6C
−4.83158
0.030174703



RAB27B
−4.81706
3.19313E−07



COBL
−4.81559
0.000878147



TMC5
−4.80912
4.21366E−11



CLGN
−4.79296
0.001549402



RAP1GAP
−4.78347
1.60818E−06



USP43
−4.77611
0.043299512



GYG2
−4.76148
0.031360574



MAP7
−4.68236
2.70581E−05



MESP1
−4.68154
0.000341157



SLC16A14
−4.63291
1.84154E−06



TMEM98
−4.6317
0.001086483



EPDR1
−4.62479
0.000150439



NIPAL1
−4.62291
0.003347226



NBEAP1
−4.61603
0.036190951



NAP1L2
−4.60869
0.01967406



ENDOD1
−4.59012
2.22009E−11



RP11-480I12.5
−4.58859
0.038581579



TMEM184A
−4.5783
0.008551888



EDA
−4.57727
0.003168575



C6orf132
−4.56958
0.034209994



MLPH
−4.56818
0.001159402



TMEM30B
−4.55692
0.001313938



CHRNA5
−4.54936
0.001615076



SOX9
−4.52975
0.015273621



PODN
−4.52946
0.008454248



RHPN2
−4.52081
0.000520944



RP11-426A6.7
−4.51391
0.004205484



FAM160A1
−4.45517
0.000145626



SHROOM1
−4.42189
1.48075E−06



PAX9
−4.39074
0.012964059



SLC1A2
−4.35607
0.030222612



ZP3
−4.35295
0.044794635



IRF6
−4.35264
7.07542E−06



PCDHB5
−4.33769
0.031523396



MARVELD3
−4.33581
0.008421801



RP11-747H7.3
−4.3352
0.017354855



LRRIQ1
−4.32898
0.047913842



SHISA6
−4.32686
0.000854246



DNAH5
−4.3124
2.06066E−05



FAM83H
−4.31012
0.027604189



APOD
−4.3051
0.032178145



CAB39L
−4.27946
4.41046E−05



GABRB3
−4.27892
0.000807785



ABCC6
−4.19027
0.016010283



ELOVL2
−4.18657
0.024445786



CLDN7
−4.16232
0.007706621



SPOCK1
−4.15854
0.007726148



EEF1A2
−4.15155
0.006294815



SYBU
−4.14684
1.47472E−06



RP11-44F14.9
−4.1427
0.035513706



ZBTB16
−4.1426
0.011262349



MANSC1
−4.1268
0.003655544



RP11-34613.4
−4.11864
0.014181168



CRISPLD1
−4.08335
0.000135526



ENPP3
−4.08046
0.009363237



SIX4
−4.07614
0.009605191



GREB1
−4.07006
0.000239832



REEP6
−4.06947
0.024043573



RPLP0P2
−4.06105
0.0047813



SIX1
−4.01941
0.04803082



OR51E1
−4.01531
0.009188572



F2RL1
−4.00093
0.003669078



DLX1
−3.99995
0.001131965



DPP4
−3.99894
0.019919664



DRAIC
−3.99642
3.34027E−05



SERINC2
−3.98847
0.027735755



PLEKHS1
−3.98619
0.006147776



PODXL2
−3.98613
0.049456799



OSR2
−3.97873
0.041789964



PRKD1
−3.97692
0.014936402



F5
−3.97154
0.000168314



RP11-255B23.3
−3.96763
0.006004447



HID1
−3.95023
0.023095214



ATP7B
−3.93918
0.014537241



CMTM4
−3.92948
0.001615076



MAPT
−3.90127
0.010183334



TACC2
−3.89662
0.008421801



BCAM
−3.87701
0.02627901



CTD-2331H7.1
−3.8732
0.010412188



TSPAN6
−3.86879
0.026094201



SLC2A12
−3.8634
0.008136488



RP11-680F20.10
−3.85507
0.030116743



WWC1
−3.85469
0.006147776



BEND4
−3.85284
0.00167161



HPGD
−3.8522
0.040413829



SGMS2
−3.83276
0.001360859



CD9
−3.83214
0.002988409



GUCY1A3
−3.83071
3.91721E−06



SORBS2
−3.80372
0.00033472



RP4-617A9.4
−3.79698
2.13211E−05



RAB25
−3.7958
0.016533393



TC2N
−3.77787
0.013369341



KIAA1324
−3.77572
0.005589822



ARHGEF26
−3.77026
0.001077207



NUPR1
−3.74028
0.003847893



MPV17L
−3.72587
0.002868759



RP11-752L20.3
−3.71907
0.005312611



STYK1
−3.71333
0.028582815



RP11-173P15.3
−3.71217
0.001627965



PCDH1
−3.67758
0.002427901



CTD-2008A1.2
−3.67627
3.99898E−05



SMPDL3B
−3.66277
0.000192708



AMACR
−3.6508
3.68572E−05



NPDC1
−3.63564
2.06066E−05



TTC39A
−3.61882
0.012515436



TMEM54
−3.61552
0.021942343



SLC38A11
−3.61052
0.034502376



DAB1
−3.6031
0.048696304



PMEPA1
−3.58662
0.002800716



FAM110B
−3.58115
0.019109222



RAB3D
−3.56739
0.001319505



KIAA1549
−3.56152
0.012964059



P3H2
−3.55822
0.017873352



RP11-588K22.2
−3.55621
0.017269032



ALDH1A3
−3.55009
0.00065603



TOM1L1
−3.52686
0.005571832



RP11-650L12.2
−3.52568
0.039535023



ILDR1
−3.52149
0.035659151



STEAP4
−3.51282
0.000119443



ZNF704
−3.51103
0.007065518



PLCB4
−3.50997
0.029596135



CREB3L4
−3.50844
0.001319505



TSPAN9
−3.47647
0.042701585



ANK3
−3.47157
7.78081E−05



RPS6KA6
−3.43505
0.029656097



PRNCR1
−3.42974
0.032653896



PDZRN3
−3.4276
0.008421801



AC027612.6
−3.42116
0.038581579



FASN
−3.41871
0.025208732



GRIP1
−3.41355
0.001456508



PPM1H
−3.39951
0.00163171



RGS2
−3.39419
0.040691341



TMEM136
−3.38738
7.17713E−05



MIPOL1
−3.3837
0.010807021



REPS2
−3.37405
0.000145626



ARHGEF37
−3.37274
0.006941479



RP11-48B3.4
−3.36568
0.019721003



CDH1
−3.35542
0.012768592



MPZL2
−3.35329
0.028809917



RP5-857K21.9
−3.33954
0.007859266



COLEC12
−3.33188
0.005759039



BAIAP2
−3.32769
0.031523396



NECTIN3
−3.32615
0.003315309



SORD
−3.31931
0.00156896



GNAI1
−3.30086
0.022933693



ALOX15
−3.30058
0.027196875



AP000689.8
−3.29912
0.016177969



SLC12A8
−3.26725
0.013595212



COL1A2
−3.25913
0.000181462



ACACA
−3.24295
0.006345868



KAZN
−3.23967
0.038516516



USP54
−3.22198
0.013128708



SLC10A5
−3.22123
0.011482303



MARVELD2
−3.21197
0.009194773



DDAH1
−3.20936
0.000804259



WNK3
−3.20931
0.019840929



MICAL2
−3.206
0.001633876



C1orf226
−3.20504
0.016804516



CXADR
−3.17825
0.012198713



TRPM4
−3.17715
0.041062369



VIPR1
−3.17145
0.007241708



SLC39A6
−3.14717
0.000348718



COL1A1
−3.13837
3.63729E−05



TBC1D30
−3.13418
0.012247234



PLPPR4
−3.125
0.028582815



TMEM56
−3.10976
0.011482303



ABCC4
−3.10395
0.000139166



CERS4
−3.10235
0.016170818



ABCA3
−3.09421
0.030503102



LAMA3
−3.07328
0.043727032



SOCS2
−3.05591
0.0279578



SLC16A1
−3.05516
0.024095115



PDGFA
−3.05248
5.06499E−05



TUB
−3.02892
0.03033514



OLFM2
−3.02779
0.030082505



RDH11
−3.02169
0.002169017



SERINC5
−3.0213
0.0068097



MT-ATP8
−3.01865
0.00653448



LGR4
−3.00892
0.003930701



RASEF
−3.00841
0.042466454



CANT1
−3.00341
0.005833148



COL5A2
−3.00162
0.000949378



GREB1L
−3.00015
0.000868222



OCLN
−2.99631
0.010988211



GPRC5C
−2.99309
0.049622084



AK4
−2.99069
0.00428589



OPHN1
−2.98333
0.029393141



SRPX2
−2.97978
0.044733512



EFNA1
−2.97167
0.024903478



REXO2
−2.97129
0.000139166



MYC
−2.96642
4.73503E−05



MPC2
−2.96398
6.81855E−05



ELOVL7
−2.9442
0.022922237



LRP11
−2.92824
0.005571832



GLRX2
−2.89483
0.000408745



NAALADL2
−2.88324
0.023804551



NECTIN4
−2.87763
0.027750065



ARHGAP28
−2.87728
0.031483355



MGST1
−2.87549
0.021267136



PRSS23
−2.86674
0.011482303



MT-ND1
−2.86064
0.000105476



MTRNR2L1
−2.85414
0.033069909



SLC9A3R2
−2.85142
0.00286168



TPD52
−2.84039
0.000888655



SLC12A2
−2.83619
0.000252516



FAM210B
−2.81462
0.000548103



FAM174B
−2.80768
0.028192137



SLC26A4
−2.80541
0.042466454



PLEKHH1
−2.79816
0.044509632



CMBL
−2.79137
0.028741604



NEO1
−2.77968
0.001893041



TPD52L1
−2.77444
0.023814858



SYTL2
−2.75795
0.002275789



ABHD11
−2.75729
0.04988753



UGDH
−2.75306
0.016533393



PPP3CA
−2.75253
0.002294732



RP5-857K21.8
−2.73877
0.006429423



CAMKK2
−2.73839
0.005597112



PCBD1
−2.73249
0.025465136



DCXR
−2.71061
0.029157884



STON1
−2.70229
0.017595961



HACD2
−2.68314
0.000641059



MALL
−2.67995
0.014548359



TRIB3
−2.677
0.011262349



TXNDC16
−2.65465
0.006307743



ENTPD5
−2.65234
0.000619674



SH3D19
−2.64312
0.017734625



MTRNR2L12
−2.62665
0.031384701



PDLIM5
−2.62535
0.006208061



MAP9
−2.61985
0.001949315



CYB561
−2.60219
0.038266611



SLC7A8
−2.59571
0.01337898



ABCB6
−2.58659
0.032581945



CD276
−2.56663
0.029848465



NBL1
−2.55827
0.049857902



STC2
−2.55729
0.030974193



THRB
−2.55343
0.009443348



FLNB
−2.53289
0.027035537



DEGS1
−2.53173
0.009443348



TRIB1
−2.51806
0.006917927



PDIA5
−2.51687
0.047522688



ITGB5
−2.49643
0.019064569



AIF1L
−2.49
0.030748717



PDE3B
−2.48754
0.008470304



NME4
−2.48399
0.009310586



SLC19A2
−2.47665
0.03778331



TMEM106C
−2.4741
0.012908923



FAM213A
−2.45981
0.022167668



PXDN
−2.45974
0.006429423



GPR160
−2.45061
0.003988421



MT-ND2
−2.44487
0.012075163



ARHGAP29
−2.44087
0.012060269



MT-ATP6
−2.43815
0.012681365



MAML3
−2.434
0.013224523



RP11-96D1.11
−2.43379
0.027908918



JUN
−2.42316
0.029656097



MT-ND4L
−2.42155
0.002012454



TRIM68
−2.41935
0.007673209



FSTL1
−2.40755
0.023289438



ENC1
−2.40286
0.042443384



SMOC2
−2.39778
0.027750065



SETD7
−2.39332
0.016010283



ZNF615
−2.39309
0.015444327



RP11-701H24.4
−2.39055
0.028582815



LCLAT1
−2.38519
0.002531903



KCTD15
−2.37194
0.016568111



MT-ND6
−2.33954
0.048113062



TRIQK
−2.33685
0.00696301



MGST2
−2.33313
0.015444327



ZBTB10
−2.33043
0.004194537



ACADSB
−2.31812
0.01333021



FKBP4
−2.3173
0.046277831



KIF21A
−2.31453
0.03778331



MTRNR2L8
−2.30359
0.026633792



NUDT19
−2.29356
0.009058911



AKAP1
−2.28996
0.019721003



ACSL3
−2.26876
0.006941479



MT-ND4
−2.24326
0.0071878



SLC25A37
−2.23936
0.020691978



NTN4
−2.23137
0.04988753



SLC7A11
−2.22381
0.016533393



PRUNE2
−2.21993
0.016533393



TOB1
−2.2117
0.038933819



IGF1R
−2.209
0.029073077



GGCT
−2.19672
0.022651032



TMED2
−2.18258
0.043013224



ERGIC1
−2.17755
0.016600723



PM20D2
−2.17005
0.017085905



GJA1
−2.16616
0.047619299



GNPNAT1
−2.16346
0.039065825



RPS24
−2.16167
0.010279784



MT-CYB
−2.1492
0.017734625



SNHG4
−2.12883
0.032665401



THBS1
−2.12064
0.010901468



NECTIN2
−2.11069
0.014537241



AC092296.1
−2.08196
0.025465136



COBLL1
−2.07932
0.049085362



MIA3
−2.07829
0.013778438



TTC7B
−2.06515
0.042152661



PUS7
−2.0614
0.037984901



AIDA
−2.05807
0.030503102



RP5-857K21.10
−2.05495
0.014508082



SGMS1
−2.04054
0.031891577



CKAP4
−2.03696
0.034778833



ATP2C1
−2.02364
0.027594022



P4HA1
−2.02335
0.038516516



MT-ND5
−2.01271
0.028362793



IARS2
−2.01144
0.032521008



LRIG1
−2.0102
0.047027728



SPARC
−2.00324
0.024869268



ATP5B
−1.99582
0.030082505



MT-RNR2
−1.98933
0.023760359



FH
−1.96081
0.029353507



CDK2AP1
−1.94144
0.046502254



MPZL1
−1.94084
0.020671596



HDLBP
−1.938
0.040525091



VKORC1L1
−1.92651
0.028536059



OCRL
−1.8641
0.049889138



IMPAD1
−1.83155
0.042443384



REEP3
−1.82779
0.038508624



CCND1
−1.78629
0.044005909

















TABLE 7







Genes upregulated in progression versus baseline in converters












log2 fold




Gene
change
p adjusted















SYNE2
1.76486
0.035090697



KLHL5
1.953712
0.043255466



SEMA4D
2.039621
0.047939776



GPCPD1
2.043504
0.024547033



ZFP36L1
2.094277
0.030147062



LMO4
2.118286
0.043806646



VAMP8
2.152774
0.025719884



MAML2
2.161568
0.013457395



SGPP2
2.172475
0.038933819



EVL
2.174487
0.03069022



ARL4C
2.289298
0.02065813



ZNF594
2.370558
0.024441058



ADGRE5
2.378299
0.037724978



TCIRG1
2.401484
0.011482303



ITPRIP
2.407407
0.043553594



ARNTL2
2.416197
0.027552182



STAB1
2.442451
0.041153521



ACSL5
2.449859
0.032607087



STARD9
2.461366
0.014178131



CNTRL
2.46191
0.045896017



C14orf159
2.468725
0.018386116



MTSS1
2.478498
0.014262051



DEF6
2.499426
0.032900387



DGKA
2.514621
0.010793965



MOXD1
2.524514
0.029953046



MEIS2
2.559851
0.047759085



PSTPIP2
2.560933
0.013128708



MCM5
2.57305
0.043299512



PKIG
2.598548
0.037724978



ARRDC2
2.602361
0.042466454



YPEL3
2.607879
0.04280022



LAT2
2.616305
0.010142873



SLCO2B1
2.620583
0.039065825



PLEKHG1
2.622
0.017336922



SHKBP1
2.627676
0.017873352



STARD4
2.64387
0.048850161



RP11-1299A16.3
2.644352
0.023760359



TNFAIP3
2.651714
0.025827419



TLR4
2.65947
0.046755101



SYT11
2.676304
0.045777071



TYMP
2.688109
0.044415341



PAQR8
2.700525
0.014508082



CDCA7L
2.714444
0.014508082



ADCY7
2.721396
0.044154503



U2AF1L4
2.725863
0.011060633



TNFAIP2
2.73051
0.003816972



TCF4
2.732309
0.000343767



ID2
2.739049
0.022651032



EPM2A
2.743283
0.038516516



AKNA
2.743529
0.042253141



FMNL1
2.754205
0.009188572



PTPN6
2.757803
0.008421801



ST6GAL1
2.78658
0.00063903



TCF7L1
2.794273
0.040070983



NAV2
2.807755
0.044005909



ITGB8
2.848144
0.011752953



UPP1
2.851644
0.029656097



SYK
2.859829
0.000248805



SLFN12
2.877584
0.028362793



CTC-479C5.12
2.900119
0.02055777



CA13
2.940601
0.044905651



PITPNC1
2.95736
0.018386116



BTN2A2
2.963709
0.01146888



RELT
2.967281
0.021267136



SIPA1
2.978846
0.025151796



VSIR
3.043285
0.032521008



CCDC88A
3.062236
0.034902216



PRSS12
3.065974
0.033794109



SEMA7A
3.069592
0.043147437



PTPRE
3.073594
0.000197808



IFI27
3.076778
0.029073077



NEDD9
3.082863
0.024449839



HOXA7
3.094877
0.047774263



RP4-530I15.9
3.108511
0.012075274



TNFRSF14
3.112997
0.00286168



ELF4
3.145261
0.006126734



APOBEC3C
3.199642
0.013229426



ESR2
3.210163
0.044005909



FAM46C
3.216277
0.000341157



HLA-DRB1
3.216307
0.042698538



APOBEC3B
3.24095
0.00654894



ATG16L2
3.263998
0.002310415



CD83
3.264546
0.019840929



ST3GAL5
3.267786
0.017269032



DOCK11
3.275624
0.005833148



FRMD3
3.278979
0.008748307



RP11-477J21.7
3.280732
0.013128708



IER5
3.283344
0.0071878



TMEM108
3.284225
0.027194057



SYNE3
3.303503
0.003024252



GAB3
3.306265
0.035832408



HMOX1
3.306551
7.52101E−05



ADORA2A
3.325043
0.011591857



LAT
3.337894
0.017085905



ISG20
3.387287
0.003315309



RP11-179F17.5
3.399836
0.037150016



HCP5
3.400245
0.006613418



ABCA7
3.404192
0.048240855



NELL2
3.407317
0.045011864



HVCN1
3.407787
0.04988753



PLA2G4A
3.410014
0.033450607



PIM2
3.420993
0.039300584



RBM38
3.425395
0.000343046



TNFSF8
3.445471
0.025371608



CIITA
3.480644
0.045400538



KYNU
3.484213
0.047759085



EGR3
3.487496
0.020111479



TMEM176B
3.489939
0.025330397



RP11-164J13.1
3.498782
0.045896017



TP63
3.505751
0.042265791



ADA
3.519396
0.000188397



NCF4
3.535008
0.040822726



CASP1
3.555885
0.038266611



TCN2
3.564498
0.022784159



RP11-705C15.3
3.569411
0.003699142



CELF2
3.571699
0.006305434



MPIG6B
3.57175
0.031891577



IGFLR1
3.572793
0.00128513



FRY
3.580113
0.016568111



RP11-705C15.2
3.588493
0.006004447



RGS18
3.603901
0.017227688



SIRPB2
3.60667
0.044406444



SLC38A1
3.609587
0.034227565



CCL5
3.634047
0.027908918



JAK3
3.647069
0.033129051



RP11-493L12.2
3.659467
0.02627901



TRIM22
3.664944
0.029649201



POU2F2
3.667907
0.01146888



APOL3
3.689628
0.04803082



SERPINB1
3.698872
0.000258057



CP
3.712217
0.001704088



ALDH1A1
3.728521
0.006821156



RP11-448G15.3
3.729897
0.03330558



IL16
3.730451
0.046150031



PLEKHA2
3.730878
0.001917887



CYBA
3.736414
0.010734603



MVP
3.738595
0.000201104



RP11-330H6.5
3.750256
0.012244921



RP11-326I11.5
3.772997
0.042253141



RP11-572O17.1
3.784717
0.025719884



RGPD1
3.792267
0.001585598



CTD-2516F10.2
3.79994
0.048963223



GIMAP1
3.805483
0.007842689



UCP2
3.806659
0.000215259



HIST1H1D
3.819261
0.034227565



ETV5
3.845065
0.000140179



CD37
3.870324
0.019522346



PLEK
3.880313
0.030503102



SCN3A
3.885338
0.043299512



ITGAL
3.913234
0.045774541



ARHGAP9
3.920188
0.04246929



CBFA2T3
3.925024
0.045258484



RP11-750H9.5
3.934689
0.039637112



CTD-2335A18.1
3.95795
0.042265791



PTPRO
3.96354
0.003841661



TTC34
3.965547
0.038216068



MIAT
3.979853
5.00879E−06



HOXA1
3.982865
0.045635625



ARSJ
3.987808
0.014537241



CXCL12
3.98824
0.029656097



XXbac-BPG299F13.17
4.000874
0.02064144



ARHGAP4
4.012078
0.000403938



CXCR3
4.021799
0.027594022



ABC12-49244600F4.3
4.023337
0.016533393



PTPRC
4.032284
0.039415496



CORO1A
4.038029
0.000765875



HLA-F
4.040062
0.019243098



WAS
4.040803
0.006429423



PARVG
4.044891
0.001755056



XCR1
4.049147
0.032521008



LIMD2
4.060399
9.26563E−06



LILRB1
4.068654
0.024445786



PPP1R16B
4.074227
0.025049208



SERPINB9
4.078595
0.013050315



KLRK1
4.084042
0.025767246



CRIP1
4.103307
0.029596135



GBP5
4.111995
0.044794635



HLA-L
4.119434
0.019674876



AC074289.1
4.119841
0.030116743



C1orf220
4.127487
0.00504242



PYGL
4.137146
4.65953E−06



GVINP1
4.14692
0.037724978



RP3-395M20.8
4.184878
0.041743802



CD200R1
4.203766
0.037518287



ADAM8
4.224928
0.001319505



NCF1C
4.226763
0.037724978



RP11-44D15.3
4.237622
0.004597535



PTPRCAP
4.250074
0.016533393



IL2RG
4.250264
0.015406855



CNTN1
4.263501
0.018865573



PTGDS
4.268047
0.025330397



GPAT2
4.270703
0.047961416



PIK3CD
4.296049
0.002299064



MSC
4.298519
0.042466454



CR1
4.302019
0.030822155



KLRD1
4.3059
0.036190951



RP11-426C22.10
4.3261
0.017438053



IKZF1
4.347873
0.028347515



S100A6
4.358014
8.39035E−05



PKD1L3
4.38512
0.005930687



CD8B
4.390215
0.011509461



TRBC1
4.417117
0.023814858



ATP2A3
4.421924
0.001266179



ARL11
4.44472
0.00175777



AMICA1
4.450267
0.030974193



CACNA1E
4.460603
0.020292054



CTB-118N6.3
4.469907
0.021713608



AC007254.3
4.477776
0.027161703



CCDC141
4.496443
0.009479687



PTHLH
4.498621
0.009550388



DAPP1
4.50607
0.030116743



GZMK
4.512197
0.009188572



CD247
4.518303
0.015768631



IL21R
4.526217
0.003312989



CD40
4.535153
0.018874524



ABCB4
4.560807
0.025719884



WDFY4
4.563862
0.000666033



IL8RBP
4.599721
0.001509204



MICB
4.612005
0.000878147



CYTIP
4.612133
0.015950048



IL24
4.613578
0.006307743



CARD11
4.615195
0.025719884



GZMH
4.618429
0.046061386



FGD2
4.637003
0.00278874



RHOH
4.640321
0.018131052



DHRS9
4.671261
0.001580651



PRF1
4.672126
0.012075274



TMEM176A
4.672206
0.001024203



IL7R
4.674792
0.008748307



HAPLN3
4.706129
0.008442538



SOCS1
4.709079
0.016854272



RP11-373L24.1
4.727501
2.13211E−05



PRTFDC1
4.733108
0.010436503



FCGR2B
4.741864
0.016003685



GPSM3
4.75138
4.28042E−06



IKZF3
4.754905
0.010816456



DGKG
4.758519
0.000145626



RP5-1171I10.5
4.765969
0.001053877



GCSAM
4.772512
0.010412188



CERKL
4.783274
0.016668672



RP11-392O17.1
4.827101
0.017992065



CD3E
4.830298
0.012860179



CD96
4.832592
0.008081538



PAX5
4.840024
0.004346743



HACD1
4.842557
0.031124974



CLEC2D
4.852645
0.009777346



TNFSF11
4.861492
0.034395134



ROBO3
4.862121
0.048367317



DMRTA1
4.862152
0.045896017



TMC8
4.87507
0.00038781



TIMD4
4.880279
0.02232107



TMEM163
4.90377
0.001585598



ITK
4.906544
0.010575746



LRMP
4.908943
0.020599231



CEMIP
4.921437
0.003886897



FSTL5
4.92619
0.025299271



MEI1
4.932381
0.022426113



PROX1
4.952832
2.13476E−11



ZNF826P
4.962978
0.029303804



RP11-358M11.2
4.965878
0.001986346



CD52
4.971075
0.002229025



GAPT
4.981553
0.027794701



NOD2
4.990199
0.000808035



HBB
5.003826
0.024445786



CD274
5.006322
0.018382137



PLCG2
5.027363
0.005303591



LINC00528
5.061849
0.032755633



RP11-61F12.1
5.075263
0.003315652



LA16c-60D12.2
5.093703
0.049085362



IRF8
5.099649
0.014413916



SLAMF1
5.106551
0.00271796



RP11-622O11.2
5.132995
0.013484008



ANO9
5.16333
0.004666712



SLAMF6
5.166919
0.020594204



AC005618.6
5.187865
0.006599332



RP11-398A8.3
5.189684
0.027061508



SLFN13
5.193984
0.021381984



KIAA0226L
5.194888
0.019923058



CD48
5.195172
0.023189266



GRAPL
5.202015
0.036190951



TRBC2
5.214755
0.006941479



KMO
5.216515
0.01699022



FPR1
5.222859
0.00060063



TDGF1
5.258904
0.003347226



KCNH8
5.270464
0.012123472



RNF183
5.287347
0.025330397



THEMIS
5.314463
7.31986E−05



LILRA1
5.316085
0.027314987



AC020951.1
5.3282
0.030441497



ZAP70
5.331009
0.006614271



IGKV1-39
5.335583
0.036732713



CD3D
5.378953
0.018040726



PRKCB
5.41952
0.001844778



SDPR
5.424391
0.008815765



CD79B
5.434795
0.02065813



RP11-500C11.3
5.446435
0.042698538



DTHD1
5.461362
0.032653896



RP11-981G7.6
5.472291
0.001225053



HK3
5.488422
0.006542022



VNN2
5.491846
0.002273087



PSTPIP1
5.506408
0.020600986



SLC2A6
5.511201
0.003315309



AC079767.4
5.516876
0.012010138



FCMR
5.527526
0.000722156



S100A8
5.538629
0.001485163



RP11-25K21.4
5.561899
0.010183334



PYHIN1
5.563373
0.004546655



CTSW
5.590185
1.83635E−05



RP5-1071N3.1
5.604375
0.039300584



GS1-410F4.2
5.614201
0.04138386



SLC9A4
5.624641
0.000879387



ZNF804A
5.63363
0.000343767



RIPOR2
5.63582
0.001704088



SNX20
5.65417
0.013451075



DTX2P1
5.655218
0.026149898



GFI1
5.655476
0.005696984



IRF4
5.676647
0.006519301



ALDH3A1
5.681413
0.042265791



CD22
5.682041
0.024185663



LY9
5.684414
0.011082838



GPR174
5.691891
0.003841661



LAX1
5.7163
0.027196875



RP11-960L18.1
5.747223
0.032581945



PDE6G
5.74771
0.043299512



TOX
5.751318
7.61414E−06



CTD-2020K17.1
5.767422
0.023420869



ENPP6
5.803448
0.025465136



P2RY10
5.811183
0.004856004



ADIRF-AS1
5.813772
0.003365526



PCDH15
5.821008
0.045258484



BCL11A
5.844474
3.17619E−05



HCST
5.853444
0.012860179



XXbac-BPG254F23.5
5.873496
0.032883497



HLA-DOB
5.947486
0.003495868



BACH2
6.002756
5.80941E−05



MAP4K1
6.042082
0.000722156



HBA1
6.046695
0.012253862



RP3-351K20.4
6.077065
0.016568111



MMP12
6.13847
0.012089491



GPR158
6.140182
0.04712138



POF1B
6.149312
0.002986091



TMEM156
6.153658
0.016533393



RP4-647C14.2
6.156388
0.037559431



RP6-99M1.2
6.194159
0.000110269



RP11-327F22.1
6.246517
0.009443348



RP11-211N8.2
6.266561
0.034227565



RP11-460N11.2
6.318951
0.013942832



SIT1
6.349488
0.000698766



TNFRSF13C
6.350606
1.95165E−10



RP11-808N1.1
6.401373
0.000457098



BNIP3P4
6.413333
0.000411631



GIMAP5
6.418087
0.002543297



C6orf141
6.440189
0.006170538



RP11-374F3.4
6.468822
0.005108237



RP1-56K13.1
6.480663
0.001319505



FCRL3
6.525075
0.000682541



IGHD
6.569965
0.046150031



NKG7
6.641471
7.09926E−06



BLK
6.664237
0.012515436



UGT8
6.669688
2.08596E−05



VPREB3
6.813197
0.021600758



NAPSB
6.85327
1.54075E−05



APOBEC3D
6.878752
 5.009E−05



TLR10
6.927124
0.003095416



CD79A
6.955214
1.06305E−05



AC104820.2
6.958632
0.01146888



CD27
6.961762
0.000343767



PARP15
6.962248
0.000589813



CD72
7.000245
3.23698E−06



PROX1-AS1
7.006641
0.016010283



CLNK
7.013311
0.039825295



POU2AF1
7.033135
0.001704088



AQP9
7.045749
1.23629E−05



CHL1
7.064701
 2.6622E−09



FAM159A
7.069956
0.002299064



AC012123.1
7.084507
0.030147062



UBD
7.085966
0.000108511



CTC-260E6.4
7.093282
0.019522346



TCL6
7.098081
0.000686703



RP11-1399P15.1
7.149517
0.046710982



RP11-374F3.5
7.173119
0.018476895



C11orf21
7.19384
0.004551851



WDR49
7.224185
0.014225089



ZC3H12D
7.236843
0.001350943



OR2I1P
7.260115
0.000158632



RP4-671O14.5
7.302321
0.00033472



RASGRF1
7.342961
0.001592099



CTC-260E6.6
7.343644
0.010427639



C10orf31
7.368453
0.004735077



CTAGE6
7.397778
0.00997142



AC090627.1
7.492274
0.006725243



SP140
7.496913
0.000166107



RP1-66N13.3
7.515983
0.043299512



IFNG
7.550884
0.04988753



PPBP
7.57855
0.01812717



FCRLA
7.596083
0.001107258



RP11-325F22.2
7.596515
0.029649201



CLEC17A
7.689823
0.029649201



CD5L
7.723779
0.010734603



RP11-445F6.2
7.754531
0.011247681



CLECL1
7.791782
0.002844035



TNFRSF13B
7.911565
0.013363493



KLRC4-KLRK1
7.97333
0.015873475



KLHL14
8.048069
0.000326698



TLR9
8.089038
0.006777866



RP11-553L6.2
8.143238
0.001291421



SPIB
8.183422
0.000120069



CD1C
8.197458
0.019064569



TIFAB
8.242314
0.011082838



AC002480.5
8.254084
0.014004416



GZMB
8.286565
0.000292658



RP11-861A13.4
8.329228
8.10582E−05



IDO1
8.513727
0.000546018



ZNF831
8.903011
0.000248055



RP11-1143G9.4
9.531873
2.43639E−07



MS4A1
9.902964
0.016533393



CCR7
10.0602
1.08457E−06



CXCL13
10.46714
0.040861189



BTLA
10.85143
0.047656045

















TABLE 8







Gene signature composition










Beltran, et al.
Zhang, et al.
Kim, et al. AR-



NEPC Up
Basal
repressed
ARG10





ASXL3
COL17A1
NRXN3
ALDH1A3


AURKA
CSMD2
ALX4
KLK3


BRINP1
CDH13
TRAF3IP2
FKBP5


C7orf76
MUM1L1
ATP2C2
KLK2


CAND2
MMP3
KDM4A
NKX3-1


DNMT1
IL33
TGFBR3
TMPRSS2


ETV5
GIMAP8
SEMA3C
PLPP1


EZH2
PDPN
RBL1
PART1


GNAO1
VSNL1
MET
PMEPA1


GPX2
BNC1
CIT
STEAP4


JAKMIP2
IGFBP7
CHAC1


KCNB2
DLK2
CABLES1


KCND2
HMGA2
FLNB


KIAA0408
NOTCH4
DAB2IP


LRRC16B
THBS2
AUTS2


MAP10
TAGLN
DAB1


MYCN
FHL1
CDC42EP4


NRSN1
ANXA8L2
CD55


PCSK1
COL4A6
TTLL3


PROX1
KCNQ5
RP11-159F24.1


RGS7
WNT7A
MYO15B


SCG3
KCNMA1
BCLAF1


SEC11C
NIPAL4
RIMS1


SEZ6
FLRT2
NEFL


SOGA3
LTBP2
GPD2


ST8SIA3
FOXI1
HPCAL4


SVOP
NGFR
SCRN1


SYT11
SERPINB13
TACC2


TRIM9
CNTNAP3B
APBB2



FGFR3
CDCA7L



ARHGAP25
GABRA5



AEBP1
MGST1



FJX1
DPF1



TNC
RAI14



MSRB3
PARP12



NRG1
PLXNA2



SERPINF1
EPB41L2



DLC1
IGSF9B



IL1A
RCOR1



DKK3
SMAD7



ERG
MAP2K6



SYNE1
FHOD3



JAG2
BIN1



JAM3
TMOD1



MRC2
SMAD6



SPARC
DUSP5



C16orf74
HUNK



FAT3
MYO10



KIRREL
CXorf57



SH2D5
SMC6



KRT6A
ARHGEF3



KRT34
STRBP



ITGA6
STXBP6



TP63
ROBO1



KRT5
TANC2



KRT14
FRMD3




GOLIM4




DPP10




WSCD1




TNFAIP2




EPHA6




SH3GL2




BCL2




BEND3




MBP




SAMD5




TMEM65




MYB




ASXL2




HRH2




KIAA0319




CREB5




AK5




PALM2-AKAP2




IKZF3




ARHGEF28









REFERENCES



  • 1. Attard, G., et al. Phase I clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. J Clin Oncol 26, 4563-4571 (2008).

  • 2. Clegg, N. J., et al. ARN-509: a novel antiandrogen for prostate cancer treatment. Cancer Res 72, 1494-1503 (2012).

  • 3. Scher, H. I., et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1-2 study. Lancet 375, 1437-1446 (2010).

  • 4. Tran, C., et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324, 787-790 (2009).

  • 5. Swami, U., McFarland, T. R., Nussenzveig, R. & Agarwal, N. Advanced Prostate Cancer: Treatment Advances and Future Directions. Trends Cancer (2020).

  • 6. Beer, T. M., et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med 371, 424-433 (2014).

  • 7. Scher, H. I., et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 367, 1187-1197 (2012).

  • 8. Armstrong, A. J., et al. ARCHES: A Randomized, Phase III Study of Androgen Deprivation Therapy With Enzalutamide or Placebo in Men With Metastatic Hormone-Sensitive Prostate Cancer. J Clin Oncol 37, 2974-2986 (2019).

  • 9. Davis, I. D., et al. Enzalutamide with Standard First-Line Therapy in Metastatic Prostate Cancer. N Engl J Med 381, 121-131 (2019).

  • 10. Abida, W., et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc Natl Acad Sci USA 116, 11428-11436 (2019).

  • 11. Annala, M., et al. Circulating Tumor DNA Genomics Correlate with Resistance to Abiraterone and Enzalutamide in Prostate Cancer. Cancer Discov 8, 444-457 (2018).

  • 12. Armstrong, A. J., et al. Prospective Multicenter Study of Circulating Tumor Cell AR-V7 and Taxane Versus Hormonal Treatment Outcomes in Metastatic Castration-Resistant Prostate Cancer. JCO Precis Oncol 4(2020).

  • 13. McKay, R. R., et al. Phase II Multicenter Study of Enzalutamide in Metastatic Castration-Resistant Prostate Cancer to Identify Mechanisms Driving Resistance. Clin Cancer Res 27, 3610-3619 (2021).

  • 14. Chen, W. S., et al. Genomic Drivers of Poor Prognosis and Enzalutamide Resistance in Metastatic Castration-resistant Prostate Cancer. Eur Urol 76, 562-571 (2019). He, M. X., et al. Transcriptional mediators of treatment resistance in lethal prostate cancer. Nat Med 27, 426-433 (2021).

  • 16. Miyamoto, D. T., et al. RNA-Seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance. Science 349, 1351-1356 (2015).

  • 17. Pal, S. K., et al. Identification of mechanisms of resistance to treatment with abiraterone acetate or enzalutamide in patients with castration-resistant prostate cancer (CRPC). Cancer 124, 1216-1224 (2018).

  • 18. Alumkal, J. J., et al. Transcriptional profiling identifies an androgen receptor activity-low, stemness program associated with enzalutamide resistance. Proc Natl Acad Sci USA 117, 12315-12323 (2020).

  • 19. Bluemn, E. G., et al. Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling. Cancer Cell 32, 474-489 e476 (2017).

  • 20. Beltran, H., et al. The role of lineage plasticity in prostate cancer therapy resistance. Clin Cancer Res (2019).

  • 21. Abdulfatah, E., et al. De novo neuroendocrine transdifferentiation in primary prostate cancer-a phenotype associated with advanced clinico-pathologic features and aggressive outcome. Med Oncol 38, 26 (2021).

  • 22. Beltran, H., et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med 22, 298-305 (2016).

  • 23. Lin, D., et al. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer Res 74, 1272-1283 (2014).

  • 24. Labrecque, M. P., et al. Molecular profiling stratifies diverse phenotypes of treatment-refractory metastatic castration-resistant prostate cancer. J Clin Invest 129, 4492-4505 (2019).

  • 25. Aggarwal, R., et al. Clinical and Genomic Characterization of Treatment-Emergent Small-Cell Neuroendocrine Prostate Cancer: A Multi-institutional Prospective Study. J Clin Oncol 36, 2492-2503 (2018).

  • 26. Alvarez, M. J., et al. Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nat Genet 48, 838-847 (2016).

  • 27. Nyquist, M. D., et al. Combined TP53 and RB1 Loss Promotes Prostate Cancer Resistance to a Spectrum of Therapeutics and Confers Vulnerability to Replication Stress. Cell Rep 31, 107669 (2020).

  • 28. Zhang, D., et al. Stem cell and neurogenic gene-expression profiles link prostate basal cells to aggressive prostate cancer. Nat Commun 7, 10798 (2016).

  • 29. Kim, D. H., et al. BET Bromodomain Inhibition Blocks an AR-Repressed, E2F1-Activated Treatment-Emergent Neuroendocrine Prostate Cancer Lineage Plasticity Program. Clin Cancer Res 27, 4923-4936 (2021).

  • 30. Ku, S. Y., et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science 355, 78-83 (2017).

  • 31. Mu, P., et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science 355, 84-88 (2017).

  • 32. Yu, J., et al. The Functional Landscape of Patient-Derived RNF43 Mutations Predicts



Sensitivity to Wnt Inhibition. Cancer Res 80, 5619-5632 (2020).

  • 33. Zhang, X., et al. Tikil is required for head formation via Wnt cleavage-oxidation and inactivation. Cell 149, 1565-1577 (2012).
  • 34. Ge, J., et al. Ubiquitin carboxyl-terminal hydrolase isozyme L5 inhibits human glioma cell migration and invasion via downregulating SNRPF. Oncotarget 8, 113635-113649 (2017).
  • 35. Rak, M. & Rustin, P. Supernumerary subunits NDUFA3, NDUFAS and NDUFA12 are required for the formation of the extramembrane arm of human mitochondrial complex I. FEBS Lett 588, 1832-1838 (2014).
  • 36. Guan, S. S., Sheu, M. L., Wu, C. T., Chiang, C. K. & Liu, S. H. ATP synthase subunit-beta down-regulation aggravates diabetic nephropathy. Sci Rep 5, 14561 (2015).
  • 37. Wang, Y., et al. Molecular events in neuroendocrine prostate cancer development. Nat Rev Urol 18, 581-596 (2021).
  • 38. Xin, Z., et al. Insulinoma-associated protein 1 is a novel sensitive and specific marker for small cell carcinoma of the prostate. Hum Pathol 79, 151-159 (2018).
  • 39. Labrecque, M. P., et al. Molecular profiling stratifies diverse phenotypes of treatment-refractory metastatic castration-resistant prostate cancer. J Clin Invest 130(2019).
  • 40. Yamashina, T., et al. Cancer stem-like cells derived from chemoresistant tumors have a unique capacity to prime tumorigenic myeloid cells. Cancer Res 74, 2698-2709 (2014).
  • 41. Lo, U. G., et al. IFNgamma-Induced IFITS Promotes Epithelial-to-Mesenchymal Transition in Prostate Cancer via miRNA Processing. Cancer Res 79, 1098-1112 (2019).
  • 42. Donehower, L. A., et al. Integrated Analysis of TP53 Gene and Pathway Alterations in The Cancer Genome Atlas. Cell Rep 28, 1370-1384 e1375 (2019).
  • 43. Chen, W. S., et al. Novel RB1-Loss Transcriptomic Signature Is Associated with Poor Clinical Outcomes across Cancer Types. Clin Cancer Res 25, 4290-4299 (2019).
  • 44. Lee, J. K., et al. N-Myc Drives Neuroendocrine Prostate Cancer Initiated from Human Prostate Epithelial Cells. Cancer Cell 29, 536-547 (2016).
  • 45. Aggarwal, R. R., et al. A Phase Ib/IIa Study of the Pan-BET Inhibitor ZEN-3694 in Combination with Enzalutamide in Patients with Metastatic Castration-resistant Prostate Cancer. Clin Cancer Res 26, 5338-5347 (2020).
  • 46. Penson, D. F., et al. Enzalutamide Versus Bicalutamide in Castration-Resistant Prostate Cancer: The STRIVE Trial. J Clin Oncol 34, 2098-2106 (2016).
  • 47. Shore, N. D., et al. Efficacy and safety of enzalutamide versus bicalutamide for patients with metastatic prostate cancer (TERRAIN): a randomised, double-blind, phase 2 study. Lancet Oncol 17, 153-163 (2016).
  • 48. Tao, D. L., et al. Molecular Testing in Patients With Castration-Resistant Prostate Cancer and Its Impact on Clinical Decision Making. JCO Precision Oncology, 1-11 (2017).
  • 49. Foye, A. & Febbo, P. G. Cancer gene profiling in prostate cancer. Methods Mol Biol 576, 293-326 (2010).
  • 50. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).
  • 51. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, 550 (2014).
  • 52. Wu, D. & Smyth, G. K. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res 40, e133 (2012).
  • 53. Lefebvre, C., et al. A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers. Mol Syst Biol 6, 377 (2010).
  • 54. Robertson, A. G., et al. Integrative Analysis Identifies Four Molecular and Clinical Subsets in Uveal Melanoma. Cancer Cell 32, 204-220 e215 (2017).
  • 55. Cejas, P., et al. Subtype heterogeneity and epigenetic convergence in neuroendocrine prostate cancer. Nat Commun 12, 5775 (2021).
  • 56. Bankhead, P., et al. QuPath: Open source software for digital pathology image analysis. Sci Rep 7, 16878 (2017).
  • 57. Grasso, C., et al. Assessing copy number alterations in targeted, amplicon-based next-generation sequencing data. J Mol Diagn 17, 53-63 (2015).
  • 58. Hovelson, D. H., et al. Rapid, ultra low coverage copy number profiling of cell-free DNA as a precision oncology screening strategy. Oncotarget 8, 89848-89866 (2017).
  • 59. Adalsteinsson, V. A., et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 8, 1324 (2017).
  • 60. Hovelson, D. H., et al. Development and validation of a scalable next-generation sequencing system for assessing relevant somatic variants in solid tumors. Neoplasia 17, 385-399 (2015).


All publications and patents mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described method and system of the disclosure will be apparent to those skilled in the art without departing from the scope and spirit of the disclosure. Although the disclosure has been described in connection with specific preferred embodiments, it should be understood that the disclosure as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the disclosure that are obvious to those skilled in the medical sciences are intended to be within the scope of the following claims.

Claims
  • 1. A method for treating prostate cancer, comprising: a) assaying a sample from a subject diagnosed with prostate cancer for the level of expression of one or more genes selected from the group consisting of RING FINGER PROTEIN 43 (RNF43), SMALL NUCLEAR RIBONUCLEOPROTEIN POLYPEPTIDE F (SNRPF), TRAB DOMAIN-CONTAINING PROTEIN 2A (TRABD2A), NADH-UBIQUINONE OXIDOREDUCTASE SUBUNIT A12 (NDUFA12), GROWTH ARREST-SPECIFIC 2-LIKE 3 (GAS2L3), RIBOSOMAL PROTEIN S24 (RPS24), DNA REPLICATION HELICASE/NUCLEASE 2 (DNA2), RETINITIS PIGMENTOSA (RP5-857K21.10), POC1 CENTRIOLAR PROTEIN B (POC1B), ADENOSINE KINASE (ADK), ATP SYNTHASE F1, SUBUNIT BETA (ATPSB), EXPORTIN, tRNA (XPOT), SOLUTE CARRIER ORGANIC ANION TRANSPORTER FAMILY, MEMBER 1B3 (SLCO1B3), and RHO-RELATED BTB DOMAIN-CONTAINING PROTEIN 1 (RHOBTB1);b) calculating a lineage plasticity score based on said level of gene expression;c) identifying subjects with a high lineage plasticity score; andd) administering a non-androgen receptor signaling inhibitor treatment to said subjects.
  • 2. The method of claim 1, wherein said treatment is an agent that blocks expression or activity of said one or more genes.
  • 3. The method of claim 1, wherein said agent is selected from the group consisting of an antibody, a nucleic acid, and a small molecule.
  • 4. A method for treating prostate cancer, comprising: a) assaying a sample from a subject diagnosed with prostate cancer for the level of expression of one or more genes selected from the group consisting of RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, and RHOBTB1;b) calculating a lineage plasticity score based on said level of gene expression;c) identifying subjects with a low lineage plasticity score; andd) administering an androgen receptor signaling inhibitor treatment to said subjects.
  • 5. The method of claim 4, wherein said treatment is enzalutamide.
  • 6. A method for measuring gene expression, comprising: a) assaying a sample from a subject having prostate cancer for the level of expression of two or more genes selected from the group consisting of RNF43, SNRPF, TRABD2A, NDUFA12, GAS2L3, RPS24, DNA2, RP5-857K21.10, POC1B, ADK, ATPSB, XPOT, SLCO1B3, and RHOBTB1;b) calculating a lineage plasticity score based on said level of gene expression.
  • 7. The method of claim 1, wherein said prostate cancer is castration-resistant prostate cancer (CRPC).
  • 8. The method of claim 1, wherein said one or more genes is two or more.
  • 9. The method of claim 1, wherein said one or more genes is five or more.
  • 10. The method of claim 1, wherein said one or more genes is all of said genes.
  • 11. The method of claim 1, wherein said sample is blood, urine, or prostate cells.
STATEMENT OF RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Patent Application No. 63/359,418, filed Jul. 8, 2022, the contents of each of which are incorporated by reference herein.

Provisional Applications (1)
Number Date Country
63359418 Jul 2022 US