Process for Measuring Tumor Response to an Initial Oncology Treatment

Information

  • Patent Application
  • 20170251973
  • Publication Number
    20170251973
  • Date Filed
    March 01, 2016
    8 years ago
  • Date Published
    September 07, 2017
    7 years ago
  • Inventors
  • Original Assignees
    • Novena Therapeutics Inc. (San Diego, CA, US)
Abstract
There is disclosed a process for determining early patient response to an initial therapy administration, comprising a rapid determination of effectiveness of a therapy after an initial treatment for a cancer indication. More specifically, the rapid determination of effectiveness is made days following initial therapy. There is further disclosed a process for determining early patient response to an initial cancer treatment, comprising (a) conducting a baseline PET FDG or PET FLT scan of the tumor region in a patient by determining tumor tissue metabolic rate and/or apoptotic rate (for FLT), (b) providing a single potentially effective dose of a therapeutic to the patient, (c) conducting a second PET FDG or PET FLT scan by determining tumor tissue metabolic rate, and (d) comparing the results of the first PET scan to the second PET scan to determine an imaging response in the PET scan results whereby at least a 1% reduction in tumor tissue metabolic rate indicates that the patient would benefit to treat the tumor with the initial cancer treatment. Preferably, in addition to the PET scans conducted before and after an initial dose of a cancer drug, the present disclosure further provides parallel before and after measurement(s) of the level of circulating tumor DNA (ctDNA) for specific oncogene mutations/alterations to identify early patient response to drug therapy.
Description
TECHNICAL FIELD

The present disclosure provides a process for determining early patient response to an initial therapy administration, comprising a rapid determination of an effectiveness of a therapy after an initial treatment for a cancer indication. More specifically, the rapid determination of effectiveness is made within one to fourteen days following initial therapy. The present disclosure further provides a process for determining early patient response to an initial cancer treatment, comprising (a) conducting a baseline PET FDG or PET FLT scan of the tumor region in a patient by determining tumor tissue metabolic rate and/or tumor apoptosis (for FLT) (b) providing a single potentially effective dose of a therapeutic to the patient, (c) conducting a second PET FUG or PET FLT scan to further determine tumor tissue metabolic rate and/or apoptosis, and (d) comparing the results of the first PET scan to the second PET scan to determine an imaging response in the PET scan results whereby at least a 1% reduction in tumor tissue metabolic rate and/or apoptosis (latter for FLT) indicates that the patient would benefit from the treatment. Preferably, in addition to the PET scans conducted before and after an initial dose of a cancer drug, the present disclosure further provides a parallel before and after measurement of the level of circulating tumor DNA (ctDNA) (found in blood or urine or other body fluids) for specific oncogene mutations/alterations to identify early patient response to drug therapy.


BACKGROUND

The standard to monitor therapy response in solid tumors has been measurements of tumor size by CT (CAT scan) or MRI, using RECIST (Response Evaluation Criteria in Solid Tumors) criteria for solid tumors, WHO criteria for lymphomas, and similar criteria for other subsets of disease. A limitation of this method is that changes in tumor size cannot be accurately assessed for many weeks and sometimes months after the initiation of tumor treatment, and therefore the patient must continue on treatment even though the patient may not be benefitting or may even be getting worse and the patient has to tolerate side effects of the treatment. The opposite can occur as well. Tumors may be improving but show an initial increase in size when measured by CAT scan or MRI or other imaging. This can occur because of many reasons including inflammatory changes. Therefore, a patient who is getting worse may stay on an ineffective treatment for months, and a patient whose tumor is getting better may have the treatment stopped.


Often, scans that measure tumor size are not repeated for 6 to 12 weeks after treatment starts because earlier scans do not reveal measurable changes. Therefore, it is routine that patients whose tumors are actually getting worse (that is, disease progression) stay on ineffective treatments for at least 6 to 12 weeks and often longer. The opposite is also true. In some patients, the treatment, as intended, causes beneficial cell death and necrosis or interferes with tumor metabolism but does not cause immediate shrinkage in tumor size. Indeed, in some cases, there can be initial growth of tumor (perhaps due to inflammation or other changes) even though a beneficial response is occurring (see Kurzrock et al Ann. Oncol. 2013 September; 24(9):2256-61. http://www.ncbi.nlm nih.gov/pubmed/23676418 and Benjamin et al. Clin. Oncol. 2007 May 1; 25(13):1760-4. http://www.ncbi.nlm nih.gov/pubmed/17470866). The latter is a well-described phenomenon in several forms of neoplasms and especially when new targeted or immunotherapy treatments are used. In these patients, the imaging repeated at 6 to 12 weeks after treatment start may indicate tumor growth and the patients may have an effective treatment stopped.


Moreover, there is also a need in the art to better segment patients for clinical trials because often highly effective therapies are missed when the patients are not better segmented for responders so as to identify patient populations who would benefit from a drug from those populations of patients who do not respond to such a drug. For example, several recombinant antibodies to insulin-like growth factor 1 (IGF-1R) showed only modest activity in larger scale clinical trials in patients with relapsed or refractory Ewing sarcoma family of tumors (ESFT). But closer observations showed a subgroup of patients who showed very significant beneficial responses to this relatively safe tumor therapy. (Chen and Sharon “IGF-1R as an anti-cancer target-trials and tribulations” Chin. J. Cancer, 2013 May; 32(5):242-52.). Therefore, there is a need in the art to identify the subgroups of patients who would benefit from a targeted cancer treatment (such as, anti-IGF-1R antibody treatment) such that subgroup effective treatments can be identified and subsequently approved with proper patient segmentation. The present disclosure was made in an effort to identify early such subgroups.


[18F] fluorodeoxyglucose (18F-FDG) uptake has been measured before and after induction chemotherapy in pediatric patients with ESFT (Ewings sarcoma) and response correlated with assessment of other types of imaging and with outcome (Hawkins et al., J. Clin. Oncol. 23:8828-8834, 2005). In another group of patients with osteosarcoma treated with chemotherapy 18F-FDG positron emission tomography (PET) correlated with histological response (Im et al., Eur. J. Nucl. Med Mol. Imaging 39:39-49, 2012). But these studies used PET imaging at traditional times, that is, after 6 weeks or months of chemotherapy.


A direct mode of action of cancer therapeutic agents often translates into meaningful measurable effects (such as, tumor shrinkage in baseline (index) lesions) after a few weeks or months of initial administration. Studies have indicated that achieving a response after the initial cycles of chemotherapy is predictive of complete remission (CR) and improved survival (von Minckwitz et al. Breast Cancer Res. 2008; 10:R30; and Hutchings et al. Blood 2006, 107:52-59.). Response criteria for solid tumors or lymphomas were developed by the WHO in an attempt to standardize the characterization of therapeutic efficacy and to facilitate comparisons between studies as well as comparisons with historical data (WHO handbook for reporting results of cancer treatment. Geneva (Switzerland): World Health Organization Offset Publication No. 48, 1979; and Miller et al., Cancer 1981, 47:207-214). Simplified and standardized response definitions for solid tumors were published by the RECIST Group in 2000 (Therasse et al., J. Natl. Cancer Inst. 2000, 92:205-216.). For cytotoxic agents, these guidelines assumed that an increase in tumor growth and/or the appearance of new lesions, usually assessed by CT scans or MRIs about eight weeks after starting the drug, and then every 8 to 12 weeks thereafter, signaled progressive disease (PD), such that the term “progression” became synonymous with drug failure. Cessation of the current treatment is recommended once PD is detected.


Increasing clinical experience indicates that traditional response criteria may not be sufficient to fully characterize activity in this new era of targeted therapies and/or biologics. For example, stable disease (SD) is characterized as either an increase or a decrease in tumor burden insufficient in magnitude to qualify as PD or a partial response (PR), respectively. With chemotherapy, SD is often transient and therefore not considered indicative of true antitumor activity. In contrast, with tyrosine kinase inhibitors (e.g., targeting epidermal growth factor receptor in non-small cell lung cancer), achieving SD has been identified as a potential surrogate end point for improved clinical outcome (median time to progression; Tsujino et al. J. Clin. Oncol. 2008; 26). Interpretation of this end point under the WHO and RECIST criteria, therefore, has been revisited in recent years, and durable modest regressions or prolonged SD achieved by these agents is, in some cases, now viewed as evidence of activity (Ratain and Eckhardt J. Clin. Oncol. 2004, 22:4442-4445).


Molecular markers are also used to better understand disease. However, these are classically utilized before treatment, and, if done later, they are repeated at the time of tumor progression on scans (many weeks or months after treatment). For instance, Lewis et al. (Modern Pathology 20:397-404, 2007) conducted a PCR test for translocation for Ewing's sarcoma for EWS-FLI1, EWS-ERG, EWS-ETV1, EWS-ETV4 and EWS-FEV as to identify appropriate translocation tumor markers. However, Lewis et al was not able to utilize the response biomarkers identified for this tumor type to be able to quickly monitor whether an initial dose of therapy can lead to tumor regression or tumor progression.


Therefore, there is a need in the art to rapidly measure the effectiveness of a cancer treatment drug or drug regimen (multiple drugs administered during a single course of therapy) in order to measure patient benefit quickly in view of the multitude of serious side effects that cancer treatment entails. This need is not only for the patient treatment situation but also for clinical trials of therapies wherein subgroups can be identified early in a treatment cycle to provide approvable criteria to determine which patients are appropriate for subgroup criteria that are within the label indications of an approved therapeutic or therapeutic combination.


RECIST defines response so RECIST is supposed to correlate with response by definition. But, there is controversy whether response correlates with survival. It may depend on the class of therapeutic, such as a lack of correlation with highly cytotoxic drugs because patient survival is compromised by toxicity. But in the situation of a targeted therapy, the correlation between a response and survival is high.


Accordingly, the main problem with current response criteria is that the patients must stay on drug for 8+ weeks even if their tumors are progressing. Assessment of response generally occurs at about 8 weeks by a scan, with reassessment about every 8 to 12 weeks thereafter. If we knew early (say on day 5) whether or not the patient was likely to respond, patients who were likely to be non-responders could move on to more effective treatments.


Even for drugs with few or no side effects, the need to be able to predict response is crucial. That is because, with traditional paradigms, response assessment is done after 8 weeks. For patients with cancer who are not responding, their tumor has been allowed to grow for those eight weeks. Current methods for prediction of response concentrate on assessment of pretreatment tumor specimens for biomarkers that might predict response to the targeted agent given. The appropriate biomarker to be used would of course vary by the agent given. And because tumors evolve and change, genuinely accurate prediction would require acquiring new tissue before each treatment, knowing the specific biomarker that is predictive for that treatment, and assaying for that biomarker. Biomarker prediction can be complicated because many patients with the predictive biomarker may still not respond, since tumors often have multiple defects, and some of the additional defects may create resistance pathways even in the presence of the predictive biomarker.


Further, the other problem with week 8 assessments of response relates to abandoned drugs (i.e., failed clinical trials). If the response rate is 15% and a tumor incidence is rare, a large randomized study is needed to prove efficacy to the FDA. But a randomized study may need to include thousands of patients to see a difference in outcome if response rates are low. The trial is likely to be negative unless very large numbers of patient are assessed because 85% of patients are, in effect, being harmed by the drug by having to take it for 8+ weeks before response is known. And once the drug is approved, if the trial can be done, most patients are not benefitting or are being harmed. Hence, there is a need for an accurate response predictor very early.


Accordingly, there is a need in the art to predict (non-pharmacokinetic) patient susceptibility to various kinase inhibitors, antibodies and other agents by actually administering a potentially effective dose rather than a radiolabeled micro-dose of the drug.


SUMMARY

The present disclosure provides a process for determining early patient response to an initial therapy administration, comprising a rapid determination of an effectiveness of a therapy after an initial treatment for a cancer indication. More specifically, the rapid determination of effectiveness is made within 1-14 days following initial therapy. The present disclosure provides a process for determining early patient response to an initial cancer treatment, comprising (a) conducting a baseline PET FDG or PET FLT scan of the tumor region in a patient for determining tumor tissue metabolic rate and/or tissue apoptosis; (b) providing a single potentially effective dose of a targeted therapeutic to the patient; (c) conducting a second PET FDG or PET FLT scan by determining tumor tissue metabolic rate; and (d) comparing the results of the first PET scan to the second PET scan to determine an imaging response in the PET scan results whereby at least a 1% reduction in tumor tissue metabolic rate indicates that the patient would benefit to treat the tumor with the initial cancer treatment. Preferably, in addition to the PET scans conducted before and after an initial dose of a cancer drug, the present disclosure further provides a parallel before and after serial measurement of the level of circulating tumor DNA (ctDNA) (measured in blood, urine or other body fluids) for specific oncogene mutations/alterations based on a biomarker specific for a tumor to identify early patient response to drug therapy.


Preferably, the PET scans determine lesion volume VOI ROIs to obtain maximum SUVmax and SUV SUV minimum SUVmin. Preferably, the present disclosure further comprises an additional urine or blood test to quantitate a cell-free DNA aberration representing a cancer marker that is conducted in conjunction with each PET scan to confirm changes caused by a single therapeutic dose of a cancer therapeutic.


Preferably, the PET scans are conducted with 2-deosy-2 [18F] fluoro D-glucose (FDG) PET scans or [18F]-fluoro-3′-deoxy-3′-L: -fluorothymidine ([18F]FLT) PET scans. Preferably, the second PET scan is conducted within 14 days after completion of the first dose of the therapeutic to the patient. Preferably, the therapeutic is a targeted therapeutic, such as a kinase inhibitor or an antibody.


The present disclosure provides a process for determining patient subgroup inclusion in a clinical trial of a targeted therapy for cancer, comprising (a) conducting a baseline PET FDG or PET FLT scan of the tumor region in a patient by determining baseline tumor tissue metabolic rate, (b) providing a single dose of a clinical trial test targeted therapeutic to the patient, (c) conducting a second PET FUG or PET FLT scan within 1-14 days following the single dose, by determining tumor tissue metabolic rate, and (d) comparing the results of the first PET scan to the second PET scan to determine an imaging response in the PET scan results, whereby at least a 1% reduction in tumor tissue metabolic rate indicates that the patient would be included in the clinical trial subgroup. Preferably, the process further comprises conducting a DNA aberration test comprising (i) conducting a baseline measurement of the level of circulating tumor DNA (ctDNA) for a specific oncogene mutations/alterations from a response biomarker specific for a tumor to identify early patient response to drug therapy, wherein the response biomarker is selected from the group consisting of the response biomarkers in Tables 1A and 1B, (ii) providing a single potentially effective dose of a therapeutic to the patient, (iii) conducting a second or serial measurement(s) of the level of circulating tumor DNA (ctDNA) for specific oncogene mutations/alterations from the response biomarker specific for a tumor to identify early patient response to drug therapy, wherein the second or serial measurements are conducted 1-14 days following the dose of the therapeutic, and (iv) comparing the results of the first measurement to subsequent measurements of ctDNA to determine that early reduction in ctDNA of at least 1% indicates that the patient would benefit to continue to treat the tumor with the therapeutic.


Preferably the response biomarker is selected from the group consisting of ABL1, BRAF, CHEK1, FANCC, GATA3, JAK2, MITF, PDCD1LG2, RBM10, STAT4, ABL2, BRCA1, CHEK2, FANCD2, GATA4, JAK3, MLH1, PDGFRA, RET. STK11, ACVR1B, BRCA2, CIC, FANCE, GATA6, JUN, MPL, PDGFRB, RICTOR, SUFU, AKT1, BRD4, CREBBP, FANCF, GID4 (C17or 39), KAT6A (MYST3), MRE11A, PDK1, RNF43, SYK, AKT2, BRIP1, CRKL, FANCG, GLI1, KDM5A, MSH2, PIK3C2B, ROS1, TAF1, AKT3, BTG1, CRLF2, FANCL, GNA11, KDM5C, MSH6, PIK3CA, RPTOR, TBX3, ALK, BTK, CSF1R, FAS, GNA13, KDM6A, MTOR, PIK3CB, RUNX1, TERC, AMER1 (FAM123B), C11 or 30 (EMSY), CTCF, FAT1, GNAQ, KDR, MUTYH, PIK3CG, RUNX1T1, TERT (promoter only), APC, CARD11, CTNNA1, FBXW7, GNAS, KEAP1, MYC, PIK3R1, SDHA, TET2, AR, CBFB, CTNNB1, FGF10, GPR124, KEL, MYCL (MYCL1), PIK3R2, SDHB, TGFBR2, ARAF, CBL, CUL3, FGF14, GRIN2A, KIT, MYCN, PLCG2, SDHC, TNFAIP3, ARFRP1, CCND1, CYLD, FGF19, GRM3, KLHL6, MYD88, PMS2, SDHD, TNFRSF14, ARID1A, CCND2, DAXX, FGF23, GSK3B, KMT2A (MLL), NF1, POLD1, SETD2, TOP1, ARID1B, CCND3, DDR2, FGF3, H3F3A, KMT2C (MLL3), NF2 POLE, SF3B1, TOP2A, ARID2, CCNE1, DICER1, FGF4, HGF, KMT2D (MLL2), NFE2L2, PPP2R1A, SLIT2, TP53, ASXL1, CD274, DNMT3A, FGF6, HNF1A, KRAS, NFKBIA, PRDM1, SMAD2, TSC1, ATM, CD79A, DOT1L, FGFR1, HRAS, LMO1, NKX2-1, PREX2, SMAD3, TSC2, ATR, CD79B, EGFR, FGFR2, HSD3B1, LRP1B, NOTCH1, PRKAR1A, SMAD4, TSHR, ATRX, CDC73, EP300, FGFR3, HSP9OAA1, LYN, NOTCH2, PRKCI, SMARCA4, U2AF1, AURKA, CDH1, EPHA3, FGFR4, IDH1, LZTR1, NOTCH3, PRKDC, SMARCB1, VEGFA, AURKB, CDK12, EPHA5, FH, IDH2, MAGI2, NPM1, PRSS8, SMO, VHL, AXIN1, CDK4, EPHA7, FLCN, IGF1R, MAP2K1, NRAS, PTCH1, SNCAIP, WISP3, AXL, CDK6, EPHB1, FLT1, IGF2, MAP2K2, NSD1, PTEN, SOCS1, WT1, BAP1, CDK8, ERBB2, FLT3, IKBKE, MAP2K4, NTRK1, PTPN11, SOX10, XPO1, BARD1, CDKN1A, ERBB3, FLT4, IKZF1, MAP3K1, NTRK2, QKI, SOX2, ZBTB2, BCL2, CDKN1B, ERBB4, FOXL2, IL7R, MCL1, NTRK3, RAC1, SOX9, ZNF217, BCL2L1, CDKN2A, ERG, FOXP1, INHBA, MDM2, NUP93, RAD50, SPEN, ZNF703, BCL2L2, CDKN2B, ERRFIL FRS2, INPP4B, MDM4, PAK3, RAD51, SPOP, BCL6, CDKN2C, ESR1, FUBP1, IRF2, MED12, PALB2, RAF1, SPTA1, BCOR, CEBPA, EZH2, GABRA6, IRF4, MEF2B, PARK2, RANBP2, SRC, BCORL1, CHD2, FAM46C, GATA1, IRS2, MEN1, PAX5, RARA, STAG2, BLM, CHD4, FANCA, GATA2, JAK1, MET, PBRM1, RB1, STATS, and combinations thereof.


Preferably the DNA aberration biomarker test that is conducted at substantially the same time as the first PET scan and the second DNA aberration biomarker test is conducted at the same time as the second PET scan. Preferably, select DNA rearrangements are also response biomarkers and are selected from the group consisting of the genes ALK, BRAF, EPOR, ETV6, IGK, JAK2, NTRK1, RAF1, ROS1, BCL2, CCND1, ETV1, EWSR1, IGL, KMT2A (MLL), PDGFRA, RARA, TMPRSS2, BCL6, CRLF2, ETV4, FGFR2, JAK1, MYC, PDGFRB, RET, TRG, BCR, EGFR, ETVS, IGH, and combinations thereof. Further, select gene fusion genes are selected from the group consisting of ABI1, CBFA2T3, EIF4A2, FUS, JAK1, MUC1, PBX1, RNF213, TET1, ABL1, CBFB, ELF4, GAS7, JAK2, MYB, PCM1, ROS1, TFE3, ABL2, CBL, ELL, GLI1, JAK3, MYC, PCSK7, RPL22, TFG, ACSL6, CCND1, ELN, GMPS, JAZF1, MYH11, PDCD1LG2 (PDL2), RPN1, TFPT, AFF1, CCND2, EML4, GPHN, KAT6A (MYST3), MYH9, PDE4DIP, RUNX1, TFRC, AFF4, CCND3, EP300, HERPUD1, KDSR, NACA, PDGFB, RUNX1T1 (ETO), TLX1, ALK, CD274 (PDL1), EPOR, HEY1, KIFSB, NBEAP1 (BCL8), PDGFRA, RUNX2, TLX3, ARHGAP26 (GRAF), CDK6, EPS15, HIP1, KMT2A (MLL), NCOA2, PDGFRB, SEC31A, TMPRSS2, ARHGEF12, CDX2, ERBB2, HIST1H4I, LASP1, NDRG1, PERI, SEPTS, TNFRSF11A, ARID1A, CHIC2, ERG, HLF, LCP1, NF1, PHF1, SEPT6, TOP1, ARNT, CHN1, ETS1, HMGA1, LMO1, NF2, PICALM, SEPT9, TP63, ASXL1, CIC, ETV1, HMGA2, LMO2, NFKB2, PIM1, SET, TPM3, ATF1, CIITA, ETV4, HOXA11, LPP, NIN, PLAG1, SH3GL1, TPM4, ATG 5, CLP1, ETV5, HOXA13, LY L 1, NOTCH1, PML, SLC1A2, TRIM24, ATIC, C LTC, ETV6, HOXA3, MAF, NPM1, POU2AF1, SNX29 (RUND-C2A), TRIP11, BCL10, CLTCL1, EWSR1, HOXA9, MAFB, NR4A3, PPP1CB, SRSF3, TTL, BCL11A, CNTRL (CEP110), FCGR2B, HOXC11, MALT1, NSD1, PRDM1, SS18, TYK2, BCL11B, COL1A1, FCRL4, HOXC13, MDS2, NTRK1, PRDM16, SSX1, USP6, BCL2, CREB3L1, FEV, HOXD11, MECOM, NTRK2, PRRX1, SSX2, WHSC1 (MMSET or NSD2), BCL3, CREB3L2, FGFR1, HOXD13, MKL1, NTRK3, PSIP1, SSX4, WHSC1L1, BCL6, CREBBP, FGFR1OP, HSP90AA1, MLF1, NUMA1, PTCH1, STAT6, YPELS, BCL7A, CRLF2, FGFR2, HSP90AB1, MLLT1 (ENL), NUP214, PTK7, STL, ZBTB16, BCL9, CSF1, FGFR3, IGH, MLLT10 (AF10), NUP98, RABEP1, SYK, ZMYM2, BCOR, CTNNB1, FLI1, IGK, MLLT3, NUTM2A, RAF1, TAF15, ZNF384, BCR, DDIT3, FNBP1, IGL, MLLT4 (AF6), OMD, RALGDS, TALI, ZNF521, BIRC3, DDX10, FOXO1, IKZF1, MLLT6, P2RY8, RAP1GDS1, TAL2, BRAF, DDX6, FOXO3, IL21R, MN1, PAFAH1B2, RARA, TBL1XR1, BTG1, DEK, FOXO4, IL3, MNX1, PAX 3, RBM15, TCF3 (E2A), C AMTA 1, DUSP22, FOXP1, IRF4, MSI2, PAX 5, RET, TCL1A (TCL1), CARS, EGFR, FSTL3, ITK, MSN, PAX 7, RHOH, TEC, and combinations thereof.


The present disclosure further provides a process for determining whether a patient would benefit for cancer treatment with a particular therapeutic, comprising (a) conducting a baseline measurement of the level of circulating tumor DNA (ctDNA) for specific oncogene mutations/alterations from a response biomarker specific for a tumor to identify early patient response to drug therapy, wherein the response biomarker is selected from the group consisting of the response biomarkers in Tables 1A and 1B, (b) providing a single potentially effective dose of a therapeutic to the patient, (c) conducting a second or serial measurement(s) of the level of circulating tumor DNA (ctDNA) for specific oncogene mutations/alterations from a response biomarker specific for a tumor, wherein the response biomarker is selected from the group consisting of the response biomarkers in Tables 1A and 1B, wherein the second or serial measurement(s) are conducted 1-14 days following the dose of the therapeutic, and (d) comparing the results of the first measurement to subsequent measurements of ctDNA to determine that early reduction in ctDNA of at least 1% indicates that the patient would benefit to continue to treat the tumor with the therapeutic. Preferably, the second or serial measurement(s) is conducted from 1-10 days after completion of a first dose of a therapeutic to the patient.





BRIEF DESCRIPTION OF THE FIGURE


FIG. 1 shows at PET scan of a person's tumor conducted prior to treatment (left) and two days after treatment (right) demonstrating early metabolic response.





DETAILED DESCRIPTION

The present disclosure provides using PET imaging in the first 14 days after treatment (and compare to pretreatment PET imaging) to be able to predict which patients will respond and who will not. This is applicable to patients with Ewing sarcoma or in general to patients with cancer treated with a variety of modalities. The ability of 18F-FDG PET imaging to be an early indicator of response and predict survival has been recently corroborated in Ewing sarcoma (Joo Hyun O1, et al. Response to early treatment evaluated with 18F-18F-FDG PET and PERCIST 1.0 predicts survival in patients with Ewing sarcoma family of tumors treated with a monoclonal antibody to the insulin-like growth factor 1 receptor. Journal of Nuclear Medicine, published on Jan. 21, 2016 as doi:10.2967/jnumed.115.162412) which is not prior art to the priority provisional patent application filed on 1 Mar. 2015.


An early marker of response, as provided herein solves several problems: (i) it permits patients who are not benefitting from therapy to be taken off that therapy and moved to another treatment after just a few days (or at most 14 days), rather than 6 weeks or months; (ii) it allows better assessment of who might be benefitting from therapy and reduces the chances that a patient with true benefit might be removed from treatment because CAT or MRI or similar imaging measuring tumor size shows “pseudogrowth” even while the tumor is dying; and (iii) it permits the early selection of responders for clinical trials such that a greater percentage of drugs will achieve statistical significance for efficacy and subsequent commercial approval with labeling requiring/suggesting that the presently disclosed procedure be conducted after a single treatment dose in order to determine who stays on the treatment and who gets moved to a different treatment. This is important because drugs that produce definitive responses, but in only small subsets of patients, may not be approvable in unselected patient populations. Yet these drugs can be successful once there is the ability to select the subgroup of responders within days of the first dose of drug.


The present disclosure provides a method for determining the effectiveness of a kinase inhibitor drug with a patient using a PET image, comprising the steps of: (1) obtaining three-dimensional images of different PET images at the time of or just prior to kinase drug administration of a therapeutic dose; (2) administering a single therapeutic dose to the, patient and (2) determining the lesion volume VOI ROIs to obtain maximum SUV/max and SUV SUV/minimum SUVmin; (3) in the same direction to cut the three-dimensional PET images obtained several faults, and each fault in the corresponding region of interest in the lesion volume VOI The lesion outline the region of interest based on a number of SUV and other activity values on the line ROI; (4) the three-dimensional PET image following the same line of the corresponding volume of the SUV activity as the reference value, the activity of drawing lines of different volumes and the corresponding Activity—volume curve; (5) calculating the time of administration of the specific activity—the area under the line of the curve of the volume; (6) in accordance with PET images acquired under different administration times to obtain and calculate different administration activity of time under the volume relationship line area under the curve, the effectiveness of drug is determined using T-test for statistical analysis.


The presently disclosed method uses early response to predict response. It is applicable to a wide range of tumors and agents, and would not require understanding the precise role of the biomarker portfolio in the tumor. Further, the use of early response biomarkers to predict tumor response, even if the overall response rate is 15% or even less, can find those patients who will be responders by giving them only one dose of drug. Therefore, in the context of oncology clinical drug trial designs, the present disclosed method to find responders 3-7 days following a single drug administration, significantly improves probability of running a successful clinical drug trial and improves overall drug approval rates.


As many as 85% of patients are harmed by a drug treatment, due to staying on an ineffective drug for eight weeks or longer before anyone realizes the drug is not effective for the patient's tumor, and having alternative perhaps more effective therapy delayed. The present method is able to determine treatment effectiveness at a much earlier time in order to change ineffective treatments earlier or to determine which patients should continue on a clinical study. For example, if 50% of the patients that are predicted to respond by the present early predictive method do respond (Joo Hyun O1 et al. Journal of Nuclear Medicine, published on Jan. 21, 2016 as doi:10.2967/jnumed.115.162412), and the overall response rate in an unselected patient population is 15%, using this method, only 15% of patients will continue on therapy unnecessarily until week 8, while using traditional imaging methods, 85% of patients will continue on treatment unnecessarily until week 8. This example assumes there are 100 patients and 15 will be true responders. Yet the present method will detect 30 patients to continue on study. The other 70 patients will move off trial after a day 5 assessment. Of the 30 patients who continue on study, 15 will respond and 15 will be unnecessarily treated until week 8+. But 15 of 100 unnecessarily treated is a lot better than 85 of 100. Further, a 50% response rate in oncology is considered a breakthrough therapy and no randomized trial is needed. The trade-off is that everyone will get a single dose of drug, but if the drug is relatively nontoxic (such as a targeted therapy) that should be acceptable. Accordingly, the present disclosed method is a non-traditional paradigm that will minimize side effect exposure when treatment benefit is not being well realized. This will also help to better design clinical drug trials such that better measurement criteria of treatment effectiveness can be utilized to better measure key risk/benefit criteria.


One technique that can be used for additional information is using FDG Positron Emission Tomography (PET) for treatment monitoring. An FDG PET (Positron Emission Tomography) scan is a radiological test that looks at tissue functioning, such as metabolic rates of tissues. With an FDG PET scan, a small amount of a radioactive sugar is injected into the blood. Cells that are growing, such as tumor cells, use sugar, and can be seen on 3-dimensional imaging. However, treatment monitoring, often comes at a later time during the course of treatment, after a patient has been exposed to multiple side effects of a treatment that may or may not have been effective. Efforts to determine particular drug effectiveness have been looking at personalized pharmacokinetic profiles. For example, uses of PET imaging has been done in combination with radiolabeled TM's (tyrosine kinase inhibitors such as erlotnib and gefitinib) to measure pharmacokinetics of the tyrosine kinase inhibitors. However, the dose of the radiolabeled TM's has been a microdose, not a full therapeutic dose. Pharmacokinetic analysis has been used to stratify patients. It was hypothesized that a patient's response to TKIs are dependent on achieving pharmacological active drug levels in tumor tissue and quantitative PET imaging can predict kinase inhibitor tumor concentrations. In this regard, VU Medical Center in the Netherlands is running a clinical trial to determine whether tumor concentrations of TKIs at pharmacological active doses can be predicted from PET studies using tracer amounts (micro-doses) of radiolabeled kinase inhibitors. However, the use of PET scans in this regard requires a correlation between local tumor drug concentration and specific efficacy.


As an additional measuring process, in addition to PET scans and run at the same time as the PET scans, a quantitative amount of circulating tumor DNA (ctDNA) in blood (plasma), urine, and other bodily fluids can be measured. What is measured is a relevant (to the tumor) biomarkers, which are specific oncogenes from cancer patients that have been shown to be correlated with radiographic (e.g., CAT, PET, MRI) indication of response to therapy. ctDNA or cell-free tumor DNA present in a cancer patient's blood (plasma) and subsequent urine that is derived from and the consequence of tumor cell death (apoptosis or necrosis) or shed in other ways. The relative amounts of ctDNA correlates with the tumor burden (i.e., number of tumor cells) within a cancer patient with higher amounts of ctDNA associated with greater tumor burden. The amount of ctDNA within a cancer patient's plasma and urine can be measured by quantitating the number of DNA copies present for specific oncogene aberrations derived from tumor cells. Therefore, as tumor burden increases in a patient, there is greater absolute number of tumor cell turnover (i.e., cell death) or other processes that shed tumor DNA, which equates to a larger number of copies of oncogenic tumor DNA present within plasma and subsequently urine. The relative measurement of ctDNA can be used to determine effectiveness of a cancer therapy and has been demonstrated for numerous metastatic cancers including breast, lung and colorectal cancer. For example, levels of the oncogene KRAS within metastatic cancer patients is correlated with responsiveness to chemotherapy. In histiocytic disorders, a cancer-related disease, approximately 50% of patients harbor a BRAF oncogene (i.e., BRAFV600E) and these patients responded to BRAF inhibitors. Relative measurement of BRAFV600E in urine from these patients receiving a BRAF inhibitor decreased at least 5-fold from baseline (prior to therapy) as compared to post-therapy and correlated with radiographic response. Furthermore, these changes in ctDNA levels are observed within days.


Comparative measurement of ctDNA levels from a single therapeutic dose at baseline and within days (1-14) post-therapy (and the serial pattern examined) is used as a responsive biomarker for determining which patients will respond to continued therapy. For example, for Ewing sarcoma patients, monitoring ctDNA levels of hybrid genes of 5′EWSR1 fused to parts of either FL11, ERG of EVT1. For instance, in lung cancer, this technique shows that an initial rise in ctDNA and then fall within days is predictive or response. In Marchetti et al. (J. Thoracic Oncl. 10:1437-1443, October 2015, and not prior art to the priority provisional application) an early prediction of a response to tyrosine kinase inhibitors by quantifying EGFR mutations (a biomarker) was found that a PCR test and sequencing of plasma found for EGFR a progressive decrease in SQ1 decrease starting from day 4 after therapy.


In conducting a biomarker analysis, the sequence of the point mutation or deletion mutation is detected with tissue (solid tumor) or plasma, urine or other body fluid samples collected before and at multiple times after (but within 14 days) the initiation of treatment. Preferably, the after samples are collected before a second dose or a second round of treatment is initiated in order to determine the effectiveness of only the single first dose or round of treatment. The initial samples (before treatment are analyzed with PCR primer sets bracketed around the cancer marker location to determine and confirm an appropriate tumor marker. For example, for EGFR mutation analysis with 42 patients, there was an exon 19 deletion in 31 cases (74%) and an L858R point mutation in exon 21 in 11 cases (26%).


Using early response to predict true response is done by comparing PET FDG at days 1 to 14, with baseline (pretherapy) PET-FDG, or comparing ctDNA levels of the biomarker on days 1 to 14, with baseline (pretherapy) ct DNA (in blood, urine or other bodily fluids) or integrating both parameters (PET-FDG and ct DNA changes). Other forms of PET scans, such as PET-FLT, may also be used.


Other molecular markers that could be used, but will be specific to the tumor to be treated, are selected from Table 1A and 1B.









TABLE 1A





INTEGRATED GENE LIST (includes mutations, amplifications, deletions and other changes)






















ABCB1
ARFRP1
AURKC
BIRC3
C11orf30
CD274
CD36
CDK2






(EMSY)
(PDL1)




ABCC1
ARID1A
AVPR2
BIRC5
C17orf39
CDX2
CD37
CDK4






(GID4)





ABCC2
ARID1B
AXIN1
BLM
CA9
CHIC2
CD38
CDK6


ABCC6
ARID2
AXL
BMI1
CAD
CHN1
CD4
CDK7


ABCG2
ASMTL
AR
BCL11A
CALCA
CLP1
CD40
CDK8


ABL1
ACSL6
ARAF
BCL3
CALCR
CLTC
CD44
CDK9


ABL2
AFF1
B2M
BCL9
CALM1
CLTCL1
CD52
CDKN1A


ACE
AFF4
BAD
BMP10
CALM2
CNTRL
CD58
CDKN1B







(CEP110)




ACPP
ARHGAP26
BAP1
BRAF
CALM3
COL1A1
CD70
CDKN2A



(GRAF)








ACTB
ARHGEF12
BARD1
BRCA1
CARD11
CREB3L1
CD74
CDKN2B


ACVR1B
ARNT
BBC3
BRCA2
CASP3
CREB3L2
CD79A
CDKN2C


ACVRL1
ATF1
BCL10
BRD4
CASP7
CSF1
CD79B
CDKN2D


ADA
ATG5
BCL11B
BRIP1
CASP8
CCNG1
CDA
CEACAM5


ADAM15
ATIC
BCL2
BRSK1
CASP9
CCR4
CDC7
CEBPA


AFP
ASXL1
BCL2A1
BTG1
CBFB
CCR5
CDC73
CENPE


AKT1
ATM
BCL2L1
BTG2
CBL
CCT6B
CDH1
CHD2


AKT2
ATP1A3
BCL2L2
BTK
CBR3
CD109
CDH2
CHD4


AKT3
ATP7A
BCL6
BTLA
CCL3
CD151
CDH20
CHEK1


ALK
ATR
BCL7A
BUB1
CCND1
CD19
CDH5
CHEK2


ALOX12
ATRX
BCOR
CAMTA1
CCND2
CD22
CDK1
CIAPIN1


ALOX12B
AURKA
BCORL1
CARS
CCND3
CD248
CDK12
CIC


AMER1
AURKB
BCR
CBFA2T3
CCNE1
CPS1
CRLF2
CSF3R


(FAM123B









or WTX)









ANGPT1
APC
BGLAP
CIITA
CLDN18
CREBBP
CSF1R
CTCF


ANGPT2
APH1A
BIRC2
CKS1B
CLU
CRKL
CSF2
CTLA4


CTNNA1
DHFR
EP300
FOXO4
FGFR2
GATA1
HOXD13
HOXD13


CTNNB1
DIABLO
EPCAM
FSTL3
FGFR3
GATA2
HBB
HSPA5


CTSG
DICER1
EPHA3
FUS
FGFR4
GATA3
HBEGF
HLF


CUL3
DKK1
EPHA5
F13B
FH
GATA4
HDAC1
HMGA1


CUX1
DLL4
EPHA6
F2
FHIT
GATA6
HDAC11
HMGA2


CXCL12
DNM2
EPHA7
F3
FLCN
GDF2
HDAC2
HOXA11


CXCR1
DNMT3A
EPHB1
F5
FLT1
GGH
HDAC4
HOXA13


CXCR2
DOT1L
EPHB4
FAF1
FLT3
GLI1
HDAC6
HOXA9


CXCR4
DPP4
EPHB6
FAM46C
FLT3LG
GLP2R
HDAC7
HOXC11


CYBA
DPYD
ERBB2
FANCA
FLT4
GNA11
HFE
HOXC13


CYLD
DTX1
ERBB3
FANCC
FLYWCH1
GNA12
HGF
HOXD11


CYP11B1
DUSP2
ERBB4
FANCD2
FOLH1
GNA13
HIF1A
IL11RA


CYP11B2
DUSP9
ERCC2
FANCE
FOLR1
GNAQ
HIST1H1C
IGH


CYP17A1
EIF4A2
ERCC5
FANCF
FOXL2
GNAS
HIST1H1D
IGK


CYP19A1
ELF4
ERG
FANCG
FOXO1
GNRHR
HIST1H1E
IGL


CYP1B1
ELL
ERRFI1
FANCL
FOXO3
GPC3
HIST1H2AC
IL3


CYP2C19
ELN
ESR1
FAS
FOXP1
GPR124
HIST1H2AG
IFNA2





(TNFRSF6)






CYP2C8
EPOR
ESR2
FASLG
FOXP4
GRIN2A
HIST1H2AL
IFNB1


CYP2C9
EPS15
ETS1
FAT1
FRS2
GRIN3B
HIST1H2AM
IFNG


CYP2D6
E2F1
ETV1
FBXO11
FUBP1
GRM3
HIST1H2BC
IGF1R


CYP3A4
EBF1
ETV4
FBXO31
FYN
GSK3B
HIST1H2BJ
IGF2


CYP4B1
ECT2L
ETV5
FBXW7
FZD1
GSTO1
HIST1H2BK
IGF2R


DDIT3
EDNRA
ETV6
FGF1
FZD10
GSTO2
HIST1H2BO
IKBKE


DDX10
EDNRB
EWSR1
FGF10
FZD2
GSTP1
HIST1H3B
IKZF1


DDX6
EED
EXOSC6
FGF14
FZD5
GTSE1
HNF1A
IKZF2


DEK
EEF2
EZH2
FGF19
FZD7
GUCY1A2
HOXA3
IKZF3


DUSP22
EGFL7
FCGR2B
FGF2
FZD8
G6PD
HPSE
IL13RA2


DAXX
EGFR
FCRL4
FGF23
GAS7
HERPUD1
HRAS
IL1A


DCT
EIF4EBP1
FEV
FGF3
GMPS
HEY1
HRH2
IL2


DDR2
ELP2
FGFR1OP
FGF4
GPHN
HIP1
HSD3B1
IL21R


DDX3X
EML4
FLI1
FGF6
GABRA6
HIST1H4I
HSP90AA1
IL25


DDX5
ENG
FNBP1
FGFR1
GADD45B
HSP90AB1
HSP90B1
IL29


ICK
JUN
LRP1B
MED12
MLLT3
NRAS
PAK3
PLA2G10


ID3
KAT6A
LRP2
MEF2B
MLLT4
NRP2
PALB2
PLA2G12A



(MYST3)


(AF6)





IDH1
KDSR
LRP6
MEF2C
MLLT6
NSD1
PARK2
PLA2G12B


IDH2
KIF5B
LRRK2
MEFV
MN1
NT5C2
PARP1
PLA2G1B


IL2RA
KMT2A
LTA
MEN1
MNX1
NTRK1
PARP2
PLA2G2A



(MLL)








IL4
KDM2B
LTF
MET
MSI2
NTRK2
PARP8
PLA2G2D


IL4R
KDM4C
LTK
MGMT
MSN
NTRK3
PASK
PLA2G2E


IL7R
KDM5A
LYN
MIB1
MUC1
NUP93
PAX5
PLA2G2F


INHBA
KDM5C
LZTR1
MITF
MYB
NUP98
PBRM1
PLA2G3


INPP4B
KDM6A
LMO2
MKI67
MYH9
NACA
PC
PLA2G5


INPP5D
KDR
MAF
MLH1
MYO18A
NBEAP1
PCBP1
PLA2G6


(SHIP)




(BCL8)




INSR
KEAP1
MAFB
MMP2
NAE1
NCOA2
PCLO
PLAU


IRF1
KEL
MAGEA1
MPL
NAMPT
NDRG1
PDCD1
PLCG1


IRF2
KIF11
MAGEA4
MRE11A
NAT2
NFKB2
PDCD11
PLCG2


IRF4
KIT
MAGED1
MS4A1
NCF2
NIN
PDCD1LG2
PLK1








(PDL2)



IRF8
KLF4
MAGI2
MSH2
NCL
NUMA1
PDGFB
PLK4


IRS2
KLHL6
MALT1
MSH3
NCOR2
NUP214
PDGFRA
PMP22


ITGA1
KLK2
MAP2K1
MSH6
NCSTN
NUTM2A
PDGFRB
PMS2


ITGA5
KLK3
MAP2K2
MST1R
NF1
NR4A3
PDK1
PNP


ITGAM
KMT2C
MAP2K4
MSTN
NF2
OMD
PDPK1
POLD1



(MLL3)








ITGAV
KMT2D
MAP3K1
MTF1
NFE2L2
OPRD1
PGF
POLE



(MLL2)








ITGB1
KRAS
MAP3K14
MTHFR
NFKB1
PAX3
PGR
POT1


ITGB2
LASP1
MAP3K6
MTOR
NFKBIA
PAX7
PHF6
PPARA


ITGB3
LCP1
MAP3K7
MTR
NGF
PBX1
PHLPP2
PPARD


ITGB5
LPP
MAPK1
MUTYH
NKX2-1
PCM1
PIK3C2B
PPARG


ITGB6
LYL1
MAPK3
MYC
NOD1
PCSK7
PIK3CA
PPP2R1A


ITK
LAG3
MCL1
MYCL
NOD2
PDE4DIP
PIK3CB
PRAME





(MYCL1)






ITPA
LEF1
MDM2
MYCN
NOTCH1
PER1
PIK3CD
PRDM1


JAZF1
LGALS1
MDM4
MYD88
NOTCH2
PHF1
PIK3CG
PREX2


JAK1
LHCGR
MED1
MYH11
NOTCH3
PICALM
PIK3R1
PRKAR1A


JAK2
LMO1
MDS2
MLF1
NPM1
P2RX7
PIK3R2
PRKCA


JAK3
LOXL2
MECOM
MLLT1
NQO1
P2RY8
PGR
PRKCB





(ENL)






JARID2
LPA
MKL1
MLLT10
NR4A1
PAG1
PIM1
PAFAH1B2





(AF10)






PLAG1
RAD51
S100A9
SSX2
SSTR2
TPM4
TNFRSF14
UBB


PML
RAD5IL3
S1PR1
SSX4
SSTR3
TRIM24
TNFRSF17
UBC


POU2AF1
RAF1
S1PR2
STL
SSTR4
TRIP11
TNFRSF4
UGT1A1


PPP1CB
RANBP2
SDHA
SLC7A11
SSTR5
TTL
TNFRSF8
UGT1A7


PRDM16
RARA
SDHB
SLCO1B1
STAT3
TEC
TNFRSF9
UMPS


PRRX1
RASGEF1A
SDHC
SLIT2
STAT4
TEK
TNFSF10
USP9X


PSIP1
RB1
SDHD
SMAD2
STAT5A
TERC
TNFSF13
VCAM1


PRKCI
RBM10
SELL
SMAD3
STAT5B
TERT
TNFSF13B
VDR


PRKDC
RELN
SELP
SMAD4
STAT6
TET1
TNKS
VEGFA


PRLR
RET
SERP2
SMARCA1
STK11
TET2
TOP1
VEGFB


PRSS8
RHEB
SERPINA1
SMARCA4
SUFU
TGFB1
TOP2A
VEGFC


PSMB5
RHOA
SERPINE1
SMARCB1
SULTIC4
TGFBR1
TOR1A
VHL


PSMB8
RICTOR
SETBP1
SMC1A
SUZ12
TGFBR2
TP53
VKORC1


PTCH1
RNF43
SETD2
SMC3
SYK
TGM2
TP63
VPS4B


PTCH2
ROCK2
SF3B1
SMC4
T
TH
TP73
VWF


PTEN
ROS1
SFTPC
SMO
TACR1
TLL2
TPMT
WHSC1









(MMSET









or









NSD2)


PTGS2
RPE65
SGK1
SNCAIP
TAF1
TLR2
TRAF2
WHSCIL1


PTH
RPS27A
SHH
SOCS1
TBL1XR1
TLR3
TRAF3
WDR90


PTK2B
RPTOR
SLC10A3
SOCS2
TBX22
TLR4
TRAF5
WISP3


PTPN11
RRM2
SLC16A1
SOCS3
TBX3
TLR5
TRPM8
WT1


PTPN2
RUNX1
SLC19A1
SOD2
TAL1
TLR7
TSC1
XBP1


PTPN6
RUNX3
SLC29A1
SOX10
TAL2
TLR8
TSC2
XIAP


(SHP-1)









PTPRC
RABEP1
SLC5A5
SOX2
TAF15
TLR9
TSHR
XPC


PTPRD
RALGDS
SLC6A2
SOX9
TCF3
TMEM30A
TUSC2
XPO1






(E2A)





PTPRO
RAP1GDS1
SEC31A
SPEN
TCL1A
TMPRSS2
TUSC3
XRCC1






(TCL1)





PRRX1
RBM15
SET
SPG7
TFE3
TMSB4XP8
TYK2
XRCC2







(TMSL3)




PSIP1
RHOH
SH3GL1
SPOP
TFG
TNC
TYMS
YPEL5


PTK7
RNF213
SLC1A2
SPP1
TFPT
TNF
TYR
YES1


QKI
RPL22
SNX29
SPTA1
TFRC
TNFAIP3
USP6
YY1AP1




(RUNDC2A)







RAC1
RPN1
SRSF3
SRC
TLX1
TNFRSF10A
U2AF1
ZBTB2


RAD21
RUNX1T1
SS18
SRSF2
TLX3
TNFRSF10B
U2AF2
ZEB2



(ETO)








RAD50
RUNX2
SSX1
SSTR1
TPM3
TNFRSF11A
UBA52
ZMYM3


ZBTB16









ZMYM2









ZNF384









ZNF521









ZNF217









ZNF24









(ZSCAN3)









ZNF703









ZRSR2
















TABLE 1B





Rearrangements and other Alterations



















ALK
FGFR1




ALK
FGFR1
PDGFRA



BCL2
FGFR2
PDGFRB



BCL6
FGFR3
RAF1



BCR
IGH
RARA



BRAF
IGK
RET



BRCA1
IGL
ROS1



BRCA2
JAK1
SYT/SSX1 SYT/SSX2



BRD4
JAK2
TMPRSS2



CCND1
KIT
TRG



CRLF2
KMT2A (MLL)




EGFR
MSH2




EPOR
MYB




ETV1
MYC




ETV4
NOTCH2




ETV5
NTRK1




ETV6
NTRK2




EWSR1
PAX3/FKHR





PAX7/FKHR




EWS-FLI





EWS/FLI1, type 1





EWS/FLI1, type 2





EWS/ERG





EWS/ETV1





EWS/ETV4





EWS/FEV









Claims
  • 1. A process for determining whether a patient would benefit from cancer treatment with a particular therapeutic, comprising (a) conducting a baseline PET scan in a patient by determining tumor tissue metabolic rate, (b) providing a single potentially effective dose of a therapeutic to the patient, (c) conducting a second PET scan of the patient by determining tumor tissue metabolic rate, wherein the second PET scan is conducted 1-14 days following the dose of the therapeutic, and (d) comparing the results of the first PET scan to the second PET scan to determine a response in the PET scan results such that at least a 1% reduction in tumor tissue metabolic rate indicates that the patient would benefit to treat the tumor with the therapeutic.
  • 2. The process for determining whether a patient would benefit for cancer treatment with a particular therapeutic of claim 1, wherein the PET scans determines lesion volume VOI ROIs to obtain maximum SUVmax and SUV SUV minimum SUVmin.
  • 3. The process for determining whether a patient would benefit from cancer treatment with a particular therapeutic of claim 1, further comprising conducting a DNA aberration marker test to quantitate a plasma-free DNA alteration of a cancer marker selected from the list of markers in Tables 1A and 1B.
  • 4. The process for determining whether a patient would benefit from cancer treatment with a particular therapeutic of claim 3, wherein the DNA aberration marker test conducted at substantially the same time as the first PET scan and the second PET scan.
  • 6. The process for determining whether a patient would benefit for cancer treatment with a particular therapeutic of claim 1, wherein the second PET scan is conducted within 14 days after completion of a first dose of a therapeutic to the patient.
  • 7. The process for determining whether a patient would benefit for cancer treatment with a particular therapeutic of claim 6, wherein the second PET scan is conducted form 1-10 days after completion of a first dose of a therapeutic to the patient.
  • 8. A process for determining patient subgroup inclusion in a clinical trial of a targeted therapy for cancer, comprising (a) conducting a baseline PET FDG or PET FLT scan of the tumor region in a patient by determining baseline tumor tissue metabolic rate, (b) providing a single dose of a clinical trial test targeted therapeutic to the patient, (c) conducting a second PET FDG or PET FLT scan within 1-14 days following the single dose, by determining tumor tissue metabolic rate, and (d) comparing the results of the first PET scan to the second PET scan to determine an imaging response in the PET scan results, whereby at least a 1% reduction in tumor tissue metabolic rate indicates that the patient would be included in the clinical trial subgroup.
  • 9. The process for determining patient subgroup inclusion in a clinical trial of a targeted therapy for cancer of claim 8, further comprising conducting a parallel before and after measurement or serial measurements of the level of circulating tumor DNA (ctDNA) for specific oncogene mutations/alterations from a response biomarker specific for a tumor to identify early patient response to drug therapy, wherein the response biomarker is selected from the group consisting of the response biomarkers in Tables 1A and 1B.
  • 10. A process for determining whether a patient would benefit for cancer treatment with a particular therapeutic, comprising (a) conducting a baseline measurement of the level of circulating tumor DNA (ctDNA) for specific oncogene mutations/alterations from a response biomarker specific for a tumor to identify early patient response to drug therapy, wherein the response biomarker is selected from the group consisting of the response biomarkers in Tables 1A and 1B, (b) providing a single potentially effective dose of a therapeutic to the patient, (c) conducting a second measurement of the level of circulating tumor DNA (ctDNA) for specific oncogene mutations/alterations from a response biomarker specific for a tumor, wherein the response biomarker is selected from the group consisting of the response biomarkers in Tables 1A and 1B, wherein the second or serial measurement(s) are conducted 1-14 days following the dose of the therapeutic, and (d) comparing the results of the first measurement to the second measurement to determine if at least a 1% reduction indicates that the patient would benefit to continue to treat the tumor with the therapeutic.
  • 11. The process for determining whether a patient would benefit for cancer treatment with a particular therapeutic of claim 10, wherein the second or serial measurement(s) are conducted from 1-10 days after completion of a first dose of a therapeutic to the patient.
CROSS REFERENCE TO RELATED APPLICATION

This patent application claims priority from U.S. provisional patent application 62/126,682 filed 1, Mar. 2015.