WO2020182995A1 - Vlcfa-containing lipids as prognostic and predictive marker for cancer therapies - Google Patents

Vlcfa-containing lipids as prognostic and predictive marker for cancer therapies Download PDF

Info

Publication number
WO2020182995A1
WO2020182995A1 PCT/EP2020/056895 EP2020056895W WO2020182995A1 WO 2020182995 A1 WO2020182995 A1 WO 2020182995A1 EP 2020056895 W EP2020056895 W EP 2020056895W WO 2020182995 A1 WO2020182995 A1 WO 2020182995A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
cpi
therapy
containing lipids
vlcfa
Prior art date
Application number
PCT/EP2020/056895
Other languages
French (fr)
Inventor
Carsten GRÜLLICH
Andreas Mock
Stefanie ZSCHÄBITZ
Original Assignee
Universität Heidelberg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universität Heidelberg filed Critical Universität Heidelberg
Publication of WO2020182995A1 publication Critical patent/WO2020182995A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/92Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving lipids, e.g. cholesterol, lipoproteins, or their receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • VLCFA-containing lipids as
  • the present invention relates to metabolic markers which are used as prognostic and/or predictive markers for a cancer therapy and/or for cancer therapy monitoring.
  • the present invention further relates to methods for the prognosis and/or therapy monitoring of cancer, wherein said methods comprise determining the metabolome in a patient sample, in particular the amount of very long-chain fatty acid-containing lipids (VLCFA-containing lipids) in the metabolome.
  • VLCFA-containing lipids very long-chain fatty acid-containing lipids
  • Metabolomics besides genomics, epigenomics, transcriptomics, and proteomics, aims to characterize and quantify metabolites in organic liquids or tissues.
  • the metabolome is influenced by both genetics and environmental factors like exercise, smoking, or nutrition.
  • Metabolomic signatures measured in serum or urine samples have been identified for various tumor entities as prognostic markers and as markers of early detection but are still not part of clinical routine Hakimi et al., 2016; Li et al, 2014; Sahu et al, 2017; Niziol et al, 2018).
  • the most commonly used technologies for measuring metabolites are mass spectroscopy (MS), enzyme-linked immunosorbent assay (ELISA), and nuclear magnetic resonance (NMR) spectroscopy. Depending on the method used, throughput, sensitivity, depth of coverage and cost vary significantly. While untargeted metabolomics describes the analysis of all measurable analytes in a given sample, targeted metabolomics is the approach of the target- oriented identification and absolute qualification of known metabolites.
  • the metabolite portfolio encompasses amino acids, vitamins, bile acids, short-, medium-, and long-chain fatty acids (FA), and lipids such as (acyl-) carnitines, glycerophospholipids (GPL), sphingolipids, or lipid precursors.
  • Deviant metabolites of tumor cells affect their microenvironment.
  • the activity and function of T cells is influenced by restriction of glucose availability or excessive lactate excretion by tumor cells (Haas et al, 2015; Lyssiotis & Kimmelman 2017).
  • CPI programmed cell death ligand 1
  • PD-L1 programmed cell death ligand 1
  • anti-PDl programmed cell death protein 1
  • anti-PD-Ll antibodies can be applied independent of risk profile in RCC and PD-L1 status in UC.
  • TMB tumor mutational burden
  • TILs tumor infiltrating lymphocytes
  • VLCFA-containing lipids very long-chain fatty acid-containing lipids
  • this object is solved by an in vitro , in vivo or ex vivo method for the prognosis and/or therapy monitoring of cancer, comprising the following steps:
  • VLCFA-containing lipids in the metabolome.
  • VLCFA-containing lipids as prognostic and predictive markers
  • the present invention provides the use of very long-chain fatty acid- containing lipids (VLCFA-containing lipids) as prognostic marker.
  • the present invention provides the use of very long-chain fatty acid- containing lipids (VLCFA-containing lipids) as predictive marker.
  • VLCFA Very long-chain fatty acids
  • LCFA Long-chain fatty acids
  • VLCFA and LCFA have been described to induce necroptosis and inflammatory signals through a variety of mechanisms (Fritsche 2015; Parisi et al., 2017). Unlike LCFA, VLCFA are not metabolized in the mitochondria but in peroxisomes, where they activate the peroxisome proliferation-activated receptor a (PPARa) signaling, controlling pivotal genes involved in lipid metabolism (Sonoda et al. , 2008).
  • PPARa peroxisome proliferation-activated receptor a
  • the present invention provides the use of VLCFA-containing lipids as prognostic marker or predictive marker for a cancer therapy and/or for cancer therapy monitoring.
  • VLCFA-containing lipids are suitable as marker for the success of a cancer therapy and/or for cancer therapy monitoring.
  • said cancer therapy is an immuno-oncological therapy.
  • Immuno-oncological treatments or therapies refer to cancer treatments whose purpose is to strengthen the body’s own immune response against cancer. The main types of
  • immunotherapy being used to treat cancer include monoclonal antibodies, immune checkpoint inhibitors, cancer vaccines and other, non-specific immunotherapies.
  • the use according to the present invention comprises response prediction to an immuno-oncologic therapy.
  • the cancer comprises locally advanced and/or metastatic cancer, preferably renal cancer and cancer of the bladder, ureters, and renal pelvis.
  • said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI).
  • CPI checkpoint inhibitors
  • said CPI treatment comprises treatment(s) targeting PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), CTLA4 (cytotoxic T-lymphocyte- associated protein 4), or LAG3 (lymphocyte-activation gene 3).
  • PD1 programmed cell death protein 1
  • PD-L1 programmeed cell death ligand 1
  • CTLA4 cytotoxic T-lymphocyte- associated protein 4
  • LAG3 lymphocyte-activation gene 3
  • CPI treatment can comprise the treatment of programmed cell death ligand 1 (PD-L1) positive tumors.
  • PD-L1 programmed cell death ligand 1
  • CPI treatment can also comprise combinations of two or more CPIs (e.g. anti-CTLA4, anti- LAG3, etc.) or combinations with other targeted cancer therapeutics such as kinase inhibitors, anti-VEGF antibodies, PPAR agonists, and/or combinations with chemotherapy.
  • CPIs e.g. anti-CTLA4, anti- LAG3, etc.
  • other targeted cancer therapeutics such as kinase inhibitors, anti-VEGF antibodies, PPAR agonists, and/or combinations with chemotherapy.
  • said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI)
  • cancer therapeutics e.g. kinase inhibitors, anti-VEGF antibodies, PPAR agonists etc.
  • the use according to the present invention preferably comprises determining the metabolome in a sample of a mammalian subject.
  • the sample preferably comprises blood, such as whole blood, plasma or serum.
  • the mammalian subject is preferably a human.
  • the metabolome is determined via mass spectrometry (MS), in particular via MS-based quantification.
  • MS mass spectrometry
  • the use is in combination with further markers
  • TMB tumor mutational burden
  • HLA human leukocyte antigen
  • tumor gene expression signatures tumor infiltrating lymphocytes and/or combinations thereof.
  • the present invention provides an in vitro , in vivo or ex vivo method for the prognosis and/or therapy monitoring of cancer.
  • Said method comprises the following steps:
  • said cancer therapy is an immuno-oncologic therapy.
  • the method is preferably for the response prediction to an immuno-oncologic therapy.
  • said cancer comprises locally advanced cancer and/or metastatic cancer, preferably renal cancer and cancer of the bladder, ureters, and renal pelvis.
  • the metabolome is preferably determined via mass spectrometry (MS), enzyme- linked immunosorbent assays (ELISA) or nuclear magnetic resonance (NMR) spectroscopy.
  • the amount of very long-chain fatty acids (VLCFA) in the metabolome is preferably determined via internal standard(s) (absolute quantification).
  • step (c) the amount of very long-chain fatty acids (VLCFA) in the metabolome is preferably determined relative to other measured metabolites in the same sample (relative quantification).
  • the method further comprises step (d):
  • step (d) grouping the VLCFA-containing lipids determined in step (b) and/or (c) into clusters
  • Said centroid of concentrations usually corresponds to the median concentration.
  • one cluster (such as named “cluster 2” below, see also e.g. Figure 3D) comprises phosphatidylcholines (PC), such as the following six: PC aa C38:0, PC aa C42:0, PC aa C42:2, PC ae C40:6, PC ae C42:3 and PC ae C44:6.
  • PC phosphatidylcholines
  • one further cluster (such as named“cluster 1” below, see also e.g. Figure 3D) comprises sphingomyelins (SM), such as the following three: SM C26: l, SM C24: l and SM (OH) C22: l.
  • SM sphingomyelins
  • centroid concentration of such a cluster can be calculated in step (e), wherein said centroid concentration corresponds to the median concentration of e.g. the six phosphatidylcholines of cluster 2 (see also e.g. Figure 3D and 3E).
  • the sample comprises blood, such as whole blood, plasma or serum; or saliva,
  • the mammalian subject is human.
  • said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI).
  • CPI treatment comprises treatment(s) targeting PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), CTLA4 (cytotoxic T-lymphocyte- associated protein 4), or LAG3 (lymphocyte-activation gene 3).
  • CPI treatment can comprise the treatment of programmed cell death ligand 1 (PD-L1) positive tumors.
  • PD-L1 programmed cell death ligand 1
  • CPI treatment can also comprise combinations of two or more CPIs (e.g. anti-CTLA4, anti- LAG3, etc.) or combinations with other targeted cancer therapeutics such as kinase inhibitors, anti-VEGF antibodies, PPAR agonists, and/or combinations with chemotherapy.
  • CPIs e.g. anti-CTLA4, anti- LAG3, etc.
  • other targeted cancer therapeutics such as kinase inhibitors, anti-VEGF antibodies, PPAR agonists, and/or combinations with chemotherapy.
  • said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI)
  • cancer therapeutics e.g. kinase inhibitors, anti-VEGF antibodies, PPAR agonists etc.
  • the method comprises determining or using further markers
  • TMB tumor mutational burden
  • HLA human leukocyte antigen
  • tumor gene expression signatures tumor infiltrating lymphocytes and/or combinations thereof.
  • CPI checkpoint inhibitors
  • the two clusters comprise sphingomyelins (SMs; cluster 1) and phosphatidylcholines (PCs; cluster 2), respectively.
  • SMs sphingomyelins
  • PCs phosphatidylcholines
  • the limit of detection was set to three times the median signal detected in the zero control samples as previously described (Tomas et al ., 2018). Metabolites were excluded when measurements were below 20% of LOD. The total number of metabolites available after data cleaning across the data set was 134 (9 acylcamitines, 21 amino acids, 10 biogenic amines, 1 hexose, 14 saturated GPLs, 14 sphingolipids, 65 unsaturated GPLs). To calculate the fraction of lipid isomers containing VLCFAs, we used the annotation of potential isobaric and isomeric lipid species measured with the AbsoluteZDO ® pi 80 kit provided by BIOCRATES Life Science AG. Before statistical analysis, measurements were log2 -transformed.
  • the study cohort encompassed 28 patients (Table 1). 25 patients (89.3%) had RCC with predominant clear cell characteristics, two patients (7.1%) had UC, and one patient was diagnosed with both UC and clear cell RCC (3.6%). With the exception of one RCC patient who was treated with atezolizumab and bevacizumab, all patients received treatment with nivolumab. Median age of our patient cohort was 64 years (range 41-79 years); 23 patients were male (82.1%) and 5 patients were female (17.9%).
  • tSNE stochastic neighbor embedding
  • Figure 2C illustrates the number of metabolites significantly associated with response and patient age stratified for the three different time points as a heatmap. Of note, the highest number of metabolites associated with response could be identified before starting the first immunotherapy dose (time point Tl).
  • uGPLs were the largest group of lipids in the data set (65 metabolites).
  • uGPLs with VLCFAs showed more correlation between each other than with uGPLs containing medium- or short-chain fatty acids (Figure 4).
  • the nine predictive VLCFA-containing lipids revealed two distinct anticorrelated clusters ( Figure 3D).
  • the first cluster comprised three sphingomyelins (SM), whereas the second cluster was composed of six phosphatidylcholines (PC).
  • SM sphingomyelins
  • PC phosphatidylcholines
  • Betof AS Nipp RD, Giobbie-Hurder A, Johnpulle RAN, Rubin K, Rubinstein SM, Flaherty KT, Lawrence DP, Johnson DB, Sullivan RJ. Impact of Age on Outcomes with
  • Fritsche KL The science of fatty acids and inflammation. Adv Nutr 2015;6:293S-301 S.
  • Lyssiotis CA Kimmelman AC. Metabolic Interactions in the Tumor Microenvironment. Trends Cell Biol 2017;27:863-75.
  • TMB tumor mutational burden
  • mRCC metastatic renal cell carcinoma

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Pathology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Biophysics (AREA)
  • Endocrinology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention relates to metabolic markers which are used as prognostic and/or predictive markers for a cancer therapy and/or for cancer therapy monitoring. The present invention further relates to methods for the prognosis and/or therapy monitoring of cancer, wherein said methods comprise determining the metabolome in a patient sample, in particular the amount of very long-chain fatty acid-containing lipids (VLCFA-containing lipids) in the metabolome.

Description

VLCFA-containing lipids as
prognostic and predictive marker for cancer therapies
The present invention relates to metabolic markers which are used as prognostic and/or predictive markers for a cancer therapy and/or for cancer therapy monitoring. The present invention further relates to methods for the prognosis and/or therapy monitoring of cancer, wherein said methods comprise determining the metabolome in a patient sample, in particular the amount of very long-chain fatty acid-containing lipids (VLCFA-containing lipids) in the metabolome.
BACKGROUND OF THE INVENTION
Metabolomics, besides genomics, epigenomics, transcriptomics, and proteomics, aims to characterize and quantify metabolites in organic liquids or tissues. The metabolome is influenced by both genetics and environmental factors like exercise, smoking, or nutrition.
There is an increasing interest in comparative metabolomic profiling for cancer research, as most published genomic and transcriptomic landscapes fell short of sufficiently explaining disease phenotypes or predicting therapy responses. The emerging hallmarks of cancer metabolism include dysregulation of glucose and amino acid uptake, opportunistic uptake of metabolites from the microenvironment, biosynthesis and NADPH production through glycolysis and citrate acid cycle, an increased need of nitrogen, and gene dysregulation caused by metabolites and other interactions with the microenvironment (Pavlova & Thompson, 2016). In fact, the phenomenon of altered glucose metabolism is routinely used to detect tumor tissues using the FDG-PET technique (Zhu et al ., 2011). Metabolomic signatures measured in serum or urine samples have been identified for various tumor entities as prognostic markers and as markers of early detection but are still not part of clinical routine Hakimi et al., 2016; Li et al, 2014; Sahu et al, 2017; Niziol et al, 2018).
The most commonly used technologies for measuring metabolites are mass spectroscopy (MS), enzyme-linked immunosorbent assay (ELISA), and nuclear magnetic resonance (NMR) spectroscopy. Depending on the method used, throughput, sensitivity, depth of coverage and cost vary significantly. While untargeted metabolomics describes the analysis of all measurable analytes in a given sample, targeted metabolomics is the approach of the target- oriented identification and absolute qualification of known metabolites. The metabolite portfolio encompasses amino acids, vitamins, bile acids, short-, medium-, and long-chain fatty acids (FA), and lipids such as (acyl-) carnitines, glycerophospholipids (GPL), sphingolipids, or lipid precursors.
As much as it is promising to study the tumor metabolism, in the age of cancer immunotherapies a special research focus has been placed on the interplay with the metabolism of immune cells, often referred to as immunometabolism. Deviant metabolites of tumor cells affect their microenvironment. For example, the activity and function of T cells is influenced by restriction of glucose availability or excessive lactate excretion by tumor cells (Haas et al, 2015; Lyssiotis & Kimmelman 2017).
Checkpoint inhibitors (CPI) have added to the treatment armamentarium in metastatic renal cell carcinoma (RCC) and urothelial cancer (UC) in the last years. While in UC the use of CPI in first line treatment has been restricted to programmed cell death ligand 1 (PD-L1) positive tumors in June 2018 following the results of the KEYNOTE-052 trial, the combination of nivolumab and ipilumumab in first line setting for intermediate and high-risk RCC has been FDA-approved and is expected to achieve EMA authorization in the near future. In second line, anti-PDl (programmed cell death protein 1) and anti-PD-Ll antibodies can be applied independent of risk profile in RCC and PD-L1 status in UC.
Positive PD-L1 expression (Teng et al, 2018), tumor mutational burden (TMB) (Yarchoan et al. , 2017), microsatellite status, and tumor infiltrating lymphocytes (TILs) (Galon et al. , 2006) are candidates for response prediction to CPI treatment. However, the predictive power of all these markers varies strongly between studies and cancer types and is also confused by differing methods and reagents used for biomarker testing.
There is emerging data that metabolomic profiles might also be predictive for treatment response to checkpoint inhibitors. In patients with melanoma and RCC treated with nivolumab, levels of kynurenine and adenosine, respectively, differed significantly between responders to CPI and non-responders (Giannakis et al. , 2017; Johnson et al. , 2016). There is a need in the art for providing improved means and methods which allow predicting the outcome or response to treatments methods of cancer, in particular immuno-oncologic treatments and therapies.
SUMMARY OF THE INVENTION
According to the present invention this object is solved by the use of very long-chain fatty acid-containing lipids (VLCFA-containing lipids) as prognostic marker or predictive marker for a cancer therapy and/or for cancer therapy monitoring.
According to the present invention this object is solved by an in vitro , in vivo or ex vivo method for the prognosis and/or therapy monitoring of cancer, comprising the following steps:
(a) providing a sample of a mammalian subject,
(b) determining the metabolome in said sample, and
(c) determining the amount of very long-chain fatty acid-containing lipids
(VLCFA-containing lipids) in the metabolome.
DESCRIPTION OF THE PREFERRED EMBODIMENTS OF THE INVENTION
Before the present invention is described in more detail below, it is to be understood that this invention is not limited to the particular methodology, protocols and reagents described herein as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. For the purpose of the present invention, all references cited herein are incorporated by reference in their entireties.
Concentrations, amounts, and other numerical data may be expressed or presented herein in a range format. It is to be understood that such a range format is used merely for convenience and brevity and thus should be interpreted flexibly to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. As an illustration, a numerical range of "1 to 21" should be interpreted to include not only the explicitly recited values of 1 to 21, but also include individual values and sub-ranges within the indicated range. Thus, included in this numerical range are individual values such as 1, 2, 3, 4, 5 .... 17, 18, 19, 20, 21 and sub-ranges such as from 2 to 10, 8 to 15, etc. This same principle applies to ranges reciting only one numerical value, such as "at least 90%". Furthermore, such an interpretation should apply regardless of the breadth of the range or the characteristics being described.
VLCFA-containing lipids as prognostic and predictive markers
As outlined above, the present invention provides the use of very long-chain fatty acid- containing lipids (VLCFA-containing lipids) as prognostic marker.
As outlined above, the present invention provides the use of very long-chain fatty acid- containing lipids (VLCFA-containing lipids) as predictive marker.
Very long-chain fatty acids (VLCFA) refer to fatty acid with 22 or more carbons.
Long-chain fatty acids (LCFA) refer to fatty acid with an aliphatic chain of 14 to 21 carbons.
VLCFA and LCFA have been described to induce necroptosis and inflammatory signals through a variety of mechanisms (Fritsche 2015; Parisi et al., 2017). Unlike LCFA, VLCFA are not metabolized in the mitochondria but in peroxisomes, where they activate the peroxisome proliferation-activated receptor a (PPARa) signaling, controlling pivotal genes involved in lipid metabolism (Sonoda et al. , 2008).
In particular, the present invention provides the use of VLCFA-containing lipids as prognostic marker or predictive marker for a cancer therapy and/or for cancer therapy monitoring.
Said VLCFA-containing lipids are suitable as marker for the success of a cancer therapy and/or for cancer therapy monitoring.
Preferably, said cancer therapy is an immuno-oncological therapy. Immuno-oncological treatments or therapies refer to cancer treatments whose purpose is to strengthen the body’s own immune response against cancer. The main types of
immunotherapy being used to treat cancer include monoclonal antibodies, immune checkpoint inhibitors, cancer vaccines and other, non-specific immunotherapies.
Preferably, the use according to the present invention comprises response prediction to an immuno-oncologic therapy.
Preferably, the cancer comprises locally advanced and/or metastatic cancer, preferably renal cancer and cancer of the bladder, ureters, and renal pelvis.
In one embodiment, said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI).
For example, said CPI treatment comprises treatment(s) targeting PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), CTLA4 (cytotoxic T-lymphocyte- associated protein 4), or LAG3 (lymphocyte-activation gene 3).
For example, CPI treatment can comprise the treatment of programmed cell death ligand 1 (PD-L1) positive tumors.
CPI treatment can also comprise combinations of two or more CPIs (e.g. anti-CTLA4, anti- LAG3, etc.) or combinations with other targeted cancer therapeutics such as kinase inhibitors, anti-VEGF antibodies, PPAR agonists, and/or combinations with chemotherapy.
In one embodiment, said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI)
(a) as CPI monotherapy,
(b) as combinations of one CPI with a second CPI, and/or
(c) as combinations of one or more CPI with other targeted cancer therapeutics (e.g. kinase inhibitors, anti-VEGF antibodies, PPAR agonists etc.) and/or
(d) as combinations of one or more CPI with chemotherapy.
The use according to the present invention preferably comprises determining the metabolome in a sample of a mammalian subject. The sample preferably comprises blood, such as whole blood, plasma or serum.
The mammalian subject is preferably a human.
In a preferred embodiment, the metabolome is determined via mass spectrometry (MS), in particular via MS-based quantification.
Further methods are enzyme-linked immunosorbent assays (ELISA) or nuclear magnetic resonance (NMR) spectroscopy
In one embodiment, the use is in combination with further markers,
preferably high PD-L1 expression, tumor mutational burden (TMB), microsatellite status, HLA (human leukocyte antigen) type, tumor gene expression signatures, and tumor infiltrating lymphocytes and/or combinations thereof.
Methods for the prognosis and/or therapy monitoring of cancer
As outlined above, the present invention provides an in vitro , in vivo or ex vivo method for the prognosis and/or therapy monitoring of cancer. -
Said method comprises the following steps:
(a) providing a sample of a mammalian subject,
(b) determining the metabolome in said sample, and
(c) determining the amount of very long-chain fatty acid-containing lipids (VLCFA-containing lipids) in the metabolome.
Preferably, said cancer therapy is an immuno-oncologic therapy.
The method is preferably for the response prediction to an immuno-oncologic therapy.
Preferably, said cancer comprises locally advanced cancer and/or metastatic cancer, preferably renal cancer and cancer of the bladder, ureters, and renal pelvis.
In step (b), the metabolome is preferably determined via mass spectrometry (MS), enzyme- linked immunosorbent assays (ELISA) or nuclear magnetic resonance (NMR) spectroscopy. In one embodiment, in step (c), the amount of very long-chain fatty acids (VLCFA) in the metabolome is preferably determined via internal standard(s) (absolute quantification).
In one embodiment, in step (c), the amount of very long-chain fatty acids (VLCFA) in the metabolome is preferably determined relative to other measured metabolites in the same sample (relative quantification).
In one embodiment, the method further comprises step (d):
(d) grouping the VLCFA-containing lipids determined in step (b) and/or (c) into clusters,
optionally further comprises step (e):
(e) calculating the centroid of concentrations for the clusters.
Said centroid of concentrations usually corresponds to the median concentration.
For example, one cluster (such as named “cluster 2” below, see also e.g. Figure 3D) comprises phosphatidylcholines (PC), such as the following six: PC aa C38:0, PC aa C42:0, PC aa C42:2, PC ae C40:6, PC ae C42:3 and PC ae C44:6.
For example, one further cluster (such as named“cluster 1” below, see also e.g. Figure 3D) comprises sphingomyelins (SM), such as the following three: SM C26: l, SM C24: l and SM (OH) C22: l.
For example, the centroid concentration of such a cluster can be calculated in step (e), wherein said centroid concentration corresponds to the median concentration of e.g. the six phosphatidylcholines of cluster 2 (see also e.g. Figure 3D and 3E).
Preferably, the sample comprises blood, such as whole blood, plasma or serum; or saliva,
Preferably, the mammalian subject is human.
In one embodiment, said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI). For example, said CPI treatment comprises treatment(s) targeting PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), CTLA4 (cytotoxic T-lymphocyte- associated protein 4), or LAG3 (lymphocyte-activation gene 3).
For example, CPI treatment can comprise the treatment of programmed cell death ligand 1 (PD-L1) positive tumors.
CPI treatment can also comprise combinations of two or more CPIs (e.g. anti-CTLA4, anti- LAG3, etc.) or combinations with other targeted cancer therapeutics such as kinase inhibitors, anti-VEGF antibodies, PPAR agonists, and/or combinations with chemotherapy.
In one embodiment, said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI)
(a) as CPI monotherapy,
(b) as combinations of one CPI with a second CPI, and/or
(c) as combinations of one or more CPI with other targeted cancer therapeutics (e.g. kinase inhibitors, anti-VEGF antibodies, PPAR agonists etc.) and/or
(d) as combinations of one or more CPI with chemotherapy.
In one embodiment, the method comprises determining or using further markers,
preferably high PD-L1 expression, tumor mutational burden (TMB), microsatellite status, HLA (human leukocyte antigen) type, tumor gene expression signatures, and tumor infiltrating lymphocytes and/or combinations thereof.
Preferred embodiments
- Abstract
The approval of checkpoint inhibitors (CPI) for a growing number of tumor entities has changed the therapeutic landscape of oncology significantly. Despite their success in a subset of patients, the majority receives an unsuccessful therapy while being exposed to the risks for an autoimmune adverse reaction. Tumor mutational burden, PD-L1 expression, and the extent of tumor-infiltrating lymphocytes have been found to be associated with successful CPI treatment, yet with inconclusive results.
We took a non-invasive metabolomic approach to understand immunotherapy response and failure in 28 urological cancer patients. In total, 134 metabolites were quantified in patient sera before the first, second, and third CPI dose. Modeling the association of metabolite concentrations with CPI response and patient characteristics revealed that all but one predictive metabolite (n=9/10) were very long-chain fatty acid-containing lipids (VLCFA- containing lipids). The best predictive performance was reached by a multivariate model of age and a centroid of VLCFA-containing lipids before the first immunotherapy (sensitivity: 0.850, specificity: 0.825, ROC: 0.935).
- Discussion
The field of immunooncology offers a fascinating new perspective for the treatment of cancer. However, the immune system is extremely complex and tightly regulated, and predictive biomarkers for CPI response are currently one of the most investigated areas of research in this field. The expression of PD-L1, the target of PD-L1/PD1 directed therapies, appears to be of limited predictive value especially in genitourinary tumors. While response rates are higher in PD-L1 positive tumors, PD-L1 negative tumors also respond to these therapies, albeit at lower rates. As another potential biomarker, TMB has been proposed because it may be a surrogate for neo-antigen expression-driven immunogenicity. However, data on TMB are still inconclusive. While a correlation with response was described in urothelial cancer for TMB- high tumors (Balar et al ., 2017), it could not be detected in renal cell cancer (Maia el al ., 2018).
The inventors took a longitudinal non-invasive metabolomic approach to reveal new insights about the mechanisms for immunotherapy response as well as to develop meaningful predictive biomarkers. They found that all but one predictive metabolite (n=9/10) were VLCFA-containing lipids.
In the present study, the best predictive performance was reached by combining patient age and a centroid of VLCFA-containing lipids before the first immunotherapy dose (sensitivity: 0.850, specificity: 0.825, ROC: 0.935). Though the numbers of patients are low, we made use of the necessary statistical means to avoid overfitting in our cohort by use of repeated 10-fold cross-validation.
The following examples and drawings illustrate the present invention without, however, limiting the same thereto. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1.
(A) Graphical abstract of the study design. Serum was collected from tumor patients before the first, second, and third dose of an immune checkpoint inhibitor therapy.
(B) A total of 134 metabolites were quantified across measurements using the Absolut e//J(/ pi 80 kit (BIOCRATES Life Sciences AG, Innsbruck, Austria).
(C) Kaplan-Meier curve of the overall survival since CPI start stratified by the response groups defined according to the best response to immunotherapy.
(C) Kaplan-Meier curve of the time of ongoing CPI treatment stratified by the response groups defined according to the best response to immunotherapy.
Figure 2.
(A) tSNE plot comparing the metabolome of all patients at the three different time points.
(B) Distribution of measured concentrations per metabolite class stratified by the three different time points.
(C) Boxplot of patient age stratified for immunotherapy response (p=0.013).
(D) Summary heatmaps illustrating the number of significant metabolites associated with response and age stratified for metabolite class (p<0.05).
Figure 3.
(A) Scatter plot of fraction of potential isobars containing VLCFAs calculated for every lipid in the data set over the -log 10 p-value of response to immunotherapy. Nine VLCFA- containing lipids were identified to be predictive at time point 1 (p<0.05).
(B) Association of these nine VLCFA-containing lipids for immunotherapy response at Tl, T2, and T3.
(C) Fraction of potential isobars with VLCFA over the length of the two fatty acids of all measured uGPLs. Linear regression fit with confidence interval (r=0.9).
(D) Correlation heatmap for the nine predictive VLCFA-containing lipids. The two clusters comprise sphingomyelins (SMs; cluster 1) and phosphatidylcholines (PCs; cluster 2), respectively.
(E) ROC statistics of predictive modeling. Figure 4. Correlation heatmap for average correlation between unsaturated glycerophospholipids (GPL) of different fatty acid lengths.
Of note the length of the two fatty acids per GPL is displayed.
EXAMPLES
EXAMPLE 1
1. Materials and Methods
1.1 Study set
Patients with metastatic RCC or UC undergoing CPI treatment at Heidelberg University Hospital were consented for biobanking of blood samples starting from 24.02.2016. Paired serum samples were collected prior to first (28 patients), second (n=18 patients), and third dose (n=16 patients) of CPI treatment, aliquoted and stored at -80°C until used. Medical records of all patients were accessed through the electronic patient charts. From the date of consent, clinical data was prospectively collected. Clinical parameters assessed included histopathologic diagnosis, tumor stage and grading, location of metastases, types and duration of and responses to prior treatment, type and duration of CPI treatment, first and best response to CPI treatment, and side effects of CPI treatment.
Overall survival (OS) was calculated from the date of first CPI dose to the date of death or date of last follow-up, respectively (last assessed on 13.02.2019). As a secondary endpoint we further included time of ongoing CPI treatment. Patients with disease control during CPI treatment (responder) were defined as achieving a complete (CR), partial (PR) remission, or stable (SD) disease. In contrast patients with a progressive disease (PD) were defined as non responders to CPI. Response was evaluated using RECIST 1.1 criteria.
1.2 Ethics Committee Approval and Patient Consent
All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. All patients provided written informed consent. Any information connected with the identity of individual subjects was excluded from further sample handling and data processing. The project was registered with the ethics committee of the University of Heidelberg (S023/2016).
1.3 Metabolomic analysis and data preprocessing Mass spectrometric metabolite quantification was performed at BIOCRATES Life Sciences AG, Innsbruck, Austria. All serum samples were analyzed with the Absolut e//J(/" pi 80 kit (BIOCRATES Life Sciences AG, Innsbruck, Austria). The kit enables the absolute quantification of 188 metabolite concentrations by using internal standards. Sample analysis was performed as previously described (Schmerler et al ., 2012) using MS technology (liquid chromatography MS for analysis of amino acids and biogenic amines and flow injection analysis MS for analysis of lipids, acylcamitines, and hexose. The limit of detection (LOD) was set to three times the median signal detected in the zero control samples as previously described (Tomas et al ., 2018). Metabolites were excluded when measurements were below 20% of LOD. The total number of metabolites available after data cleaning across the data set was 134 (9 acylcamitines, 21 amino acids, 10 biogenic amines, 1 hexose, 14 saturated GPLs, 14 sphingolipids, 65 unsaturated GPLs). To calculate the fraction of lipid isomers containing VLCFAs, we used the annotation of potential isobaric and isomeric lipid species measured with the AbsoluteZDO® pi 80 kit provided by BIOCRATES Life Science AG. Before statistical analysis, measurements were log2 -transformed.
1.4 Statistical analysis
All statistical analyses were conducted within the R software environment (R version 3.4.2; www.r-project.org). The data was stored within a ' SummarizedExperimenf class object enabling coordinated representation of the metabolomic data and associated metadata. Heatmaps were illustrated using the ComplexHeatmap package and boxplots and scatter plots using the ggplot2 package. T-distributed stochastic neighbor embedding (tSNE) was performed using the Rtsne R package. To determine metabolites associated with therapy response, we employed multivariate linear mixed effects modelling. Linear mixed effects models (LMMs) extend the modelling of fixed effects (therapy response and patient age) by random effects (between patient variation). The nlme package offers the functionality of LMMs within R (Pinheiro 2009). In our predictive modelling, we compared the predictive performance of patient age and the median concentration (i.e. the centroids) of the VLCFA- containing lipids belonging to cluster 1 and cluster 2 (compare figure 2D) in univariate and multivariate models. To compensate for overfitting, we applied 10-fold cross-validation, which was repeated 10 times, for each model. In addition, we used a linear (logistic regression) and a non-linear (support vector machine; svm) approach. The predictive modelling was performed with the caret R package. All presented p-values are multiplicity unadjusted. A p-value <0.05 was considered significant. 2. Results
2.1 Study design and patient cohort
Serum samples were collected from tumor patient before the first, second, and third dose of an immune checkpoint inhibitor (time points Tl, T2, T3; Figure 1 A). A total of 134 metabolites were quantified across measurements (Figure IB). The study cohort encompassed 28 patients (Table 1). 25 patients (89.3%) had RCC with predominant clear cell characteristics, two patients (7.1%) had UC, and one patient was diagnosed with both UC and clear cell RCC (3.6%). With the exception of one RCC patient who was treated with atezolizumab and bevacizumab, all patients received treatment with nivolumab. Median age of our patient cohort was 64 years (range 41-79 years); 23 patients were male (82.1%) and 5 patients were female (17.9%). Best response to treatment was PR in 4 patients (14.3%), SD in 8 patients (28.6%), and PD in 15 patients (53.6%). No CR was observed. The overall survival since CPI initiation was significantly higher in responders to CPI (Figure 1C). The same observation hold true for the time of ongoing CPI treatment (Figure ID).
Table 1. Clinicopathological characteristics of patient cohort.
Variable Total [%]
Sex
Male 23 [82.1]
Female 5 [17.9]
Median age [Range] 64 [41-79]
Tumor type
Clear cell renal cell carcinoma 25 [89.3]
Transitional cell carcinoma 2 [7.1]
Transitional cell carcinoma and 1 [3.6]
renal cell carcinoma
Site of metastases
Lung 19 [67.9]
Bone 11 [39.3]
Liver 9 [32.1]
Brain 4 [14.3]
Pancreas 4 [14.3]
Peritoneum 5 [17.9]
Lymph nodes 13 [46.4]
Other 14 [42.9]
Type of treatment
Nivolumab 27 [96.4]
Atezolizumab + Bevacizumab 1 [3.6]
Response to first staging
Partial response 3 [10.7] Stable disease 8 [8,6]
Progressive di sease 16 [57.1 ]
Missing data 1 [3.6]
Best response to treatment
Partial response 4 [14.3]
Stable disease 8 [28.6]
Progressive disease 15 [53.6]
Missing data 1 [3.6]
2.2 Unsupervised analysis of serum metabolomes
As a first investigation, we visualized the metabolome-wide variance in the data set by means of t-distributed stochastic neighbor embedding (tSNE). The tSNE plot revealed greater variance in metabolomic measurements between patients than within the same patient over time (Figure 2 A). The 134 metabolites analyzed were assigned to one of seven metabolite classes (hexoses, amino acids, sphingolipids, unsaturated GPLs, saturated GPLs, biogenic amines, and acylcamitines). Calculating the average concentration for these classes showed that the pool of metabolites remained constant over the three measured time points (Figure 2B).
2.3 Metabolites associated with response to immunotherapy
Before comparing the metabolomes of immunotherapy responders and non-responders, we considered known prognostic factors in our patient cohort. In line with published studies (Betof et al ., 2017), we could identify responders to immunotherapy to be significantly younger than non-responders (Figure 2C). Hence, we applied a multivariate modelling approach revealing differentially abundant metabolites in the response groups independent of patient age. Figure 2D illustrates the number of metabolites significantly associated with response and patient age stratified for the three different time points as a heatmap. Of note, the highest number of metabolites associated with response could be identified before starting the first immunotherapy dose (time point Tl).
2.4 Very long-chain fatty acid-containing lipids are predictive for immunotherapy response
Intriguingly, all but one metabolite (n=9/10) associated with response to immunotherapy at Tl were very long-chain fatty acid-containing lipids (VLCFA-containing lipids; Table 2). This observation became especially clear when plotting the fraction of potential isomers containing VLCFAs for every lipid over the response p-value (Figure 3 A). In addition, we could observe that their predictive performance continuously decreased from Tl to T3 (Figure 3B). To corroborate the role of VLCFAs, we compared the fraction of potential VLCFA- containing lipids with the length of the two fatty acids of all unsaturated GPLs (uGPLs), yielding a strong correlation (Figure 3C, r=0.90). We used uGPLs for this comparison, as they were the largest group of lipids in the data set (65 metabolites). In addition, uGPLs with VLCFAs showed more correlation between each other than with uGPLs containing medium- or short-chain fatty acids (Figure 4). Interestingly, the nine predictive VLCFA-containing lipids revealed two distinct anticorrelated clusters (Figure 3D). The first cluster comprised three sphingomyelins (SM), whereas the second cluster was composed of six phosphatidylcholines (PC).
Table 2. Metabolites associated with immunotherapy response independent of patient age (p<0.05). å FA chain length = sum of fatty acid chain length; isomers > 22C=fraction of potential isomeric lipids containing at least one fatty acid with a chain length greater than or equal to 22; sGPL=saturated glycerophospholipid; uGPL=unsaturated glycerophospholipid; PC=phosphatidylcholine; SM=sphingomyelin. *PC aa C38:0 has two potential isomers with unsaturated fatty acids. log2 (fold-
Figure imgf000016_0001
A chain isomers >
Metabolite class change) p-value length 22C
PC aa C38:0* sGPL 0.488128877 0.027 38 7/12
PC aa C42:0 sGPL 0.566755571 0.031 42 3/4
PC aa C42:2 uGPL 0.437668994 0.013 42 6/6
PC ae C40:6 uGPL 0.420863679 0.047 40 4/6
PC ae C42:3 uGPL 0.615033724 0.005 42 6/8
PC ae C44:6 uGPL 0.512514563 0.024 44 2/2
SM (OH)
C22: l sphingolipid 0.444489484 0.047 22 4/4
SM C24: l sphingolipid 0.461333761 0.025 24 2/2
SM C26: l sphingolipid 0.517973234 0.029 26 1/1
SM C20:2 sphingolipid 0.4872320 0.039 20 0/2
2.5 Predictive modeling of therapy response
Due to the lack of an independent validation cohort, we performed rigorous predictive modelling in our patient set to estimate the predictive performance of the VLFCA-containing lipids. To compensate for overfitting, we applied 10-fold cross-validation, which was repeated 10 times, for each model. In addition, we used a linear (logistic regression) and a non-linear (support vector machine; svm) approach. The best performance was reached by a multivariate logistic regression model including age and the centroid 2, corresponding to the median concentration of the six phosphatidylcholines presented in cluster 2 of Figure 3D (sensitivity: 0.850, specificity: 0.825, ROC: 0.935; Figure 3E, Table 3).
Table 3. ROC statistics of predictive modeling svm: support vector machine.
Formula Statistic _ ROC Sensitivity Specificity age logistic regression 0.77125 0.805 0.645 centroid 1 logistic regression 0.6825 0.805 0.54 centroid2 logistic regression 0.79 0.805 0.64 age + centroid 1 logistic regression 0.825 0.815 0.64 age + centroid2 logistic regression 0.935 0.85 0.825 age + centroid 1 + centroid2 logistic regression 0.865 0.82 0.73 age svm 0.48375 0.89 0.115 centroid 1 svm 0.4225 0.845 0.09 centroid2 svm 0.6825 0.85 0.275 age + centroid 1 svm 0.725 0.79 0.46 age + centroid2 svm 0.78 0.79 0.65 age + centroid 1 + centroid2 svm 0.8925 0.81 0.75
REFERENCES
Balar AV, Castellano D, O'Donnell PH, Grivas P, Vuky J, Powles T, Plimack ER, Hahn NM, de Wit R, Pang L, Savage MJ, Perini RF, et al. First-line pembrolizumab in cisplatin- ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 2017; 18: 1483-92.
Betof AS, Nipp RD, Giobbie-Hurder A, Johnpulle RAN, Rubin K, Rubinstein SM, Flaherty KT, Lawrence DP, Johnson DB, Sullivan RJ. Impact of Age on Outcomes with
Immunotherapy for Patients with Melanoma. Oncologist 2017;22:963-71.
Fritsche KL. The science of fatty acids and inflammation. Adv Nutr 2015;6:293S-301 S.
Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, Tosolini M, Camus M, Berger A, Wind P, Zinzindohoue F, Bruneval P, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science
2006;313 : 1960-4. Giannakis M, Li H, Jin C, Gopal S, Desai K, Horak C, Wind-Rotolo M, Van Allen EM, Clish C, Hodi FS, Garraway LA, Choueiri TK. Metabolomic correlates of response in nivolumab- treated renal cell carcinoma and melanoma patients. J Clin Oncol 2017;35:3036-6.
Haas R, Smith J, Rocher-Ros V, Nadkarni S, Montero-Melendez T, D'Acquisto F, Bland EJ, Bombardieri M, Pitzalis C, Perretti M, Marelli-Berg FM, Mauro C. Lactate Regulates Metabolic and Pro-inflammatory Circuits in Control of T Cell Migration and Effector Functions. PLoS Biol 2015 ; 13 :e 1002202.
Hakimi AA, Reznik E, Lee C-H, Creighton CJ, Brannon AR, Luna A, Aksoy BA, Liu EM, Shen R, Lee W, Chen Y, Stirdivant SM, et al. An Integrated Metabolic Atlas of Clear Cell Renal Cell Carcinoma. Cancer Cell 2016;29: 104-16.
Johnson CH, Spilker ME, Goetz L, Peterson SN, Siuzdak G. Metabolite and Microbiome Interplay in Cancer Immunotherapy. Cancer Res 2016;76:6146-52.
Li B, Qiu B, Lee DSM, Walton ZE, Ochocki JD, Mathew LK, Mancuso A, Gade TPF, Keith B, Nissim I, Simon MC. Fructose- 1,6-bisphosphatase opposes renal carcinoma progression. Nature 2014;513:251-5.
Lyssiotis CA, Kimmelman AC. Metabolic Interactions in the Tumor Microenvironment. Trends Cell Biol 2017;27:863-75.
Maia MC, Almeida L, Bergerot PG, Dizman N, Pal SK. Relationship of tumor mutational burden (TMB) to immunotherapy response in metastatic renal cell carcinoma (mRCC). J Clin Oncol 2018;36:662-2.
Niziol J, Bonifay V, Ossolihski K, Ossolihski T, Ossolihska A, Sunner J, Beech I,
Arendowski A, Ruman T. Metabolomic study of human tissue and urine in clear cell renal carcinoma by LC-HRMS and PLS-DA. Anal Bioanal Chem 2018;410:3859-69.
Parisi LR, Li N, Atilla-Gokcumen GE. Very Long Chain Fatty Acids Are Functionally Involved in Necroptosis. Cell Chem Biol 2017;24: 1445-8. Pavlova NN, Thompson CB. The Emerging Hallmarks of Cancer Metabolism. CellMetab 2016;23:27-47.
Pinheiro J. nlme : Linear and Nonlinear Mixed Effects Models. http://cranr- projectorg/web/packages/nlme/indexhtml 2009.
Sahu D, Lotan Y, Wittmann B, Neri B, Hansel DE. Metabolomics analysis reveals distinct profiles of nonmuscle-invasive and muscle-invasive bladder cancer. Cancer Med
2017;6:2106-20.
Schmerler D, Neugebauer S, Ludewig K, Bremer- Streck S, Brunkhorst FM, Kiehntopf M. Targeted metabolomics for discrimination of systemic inflammatory disorders in critically ill patients. J Lipid Res 2012;53: 1369-75.
Sonoda J, Pei L, Evans RM. Nuclear receptors: decoding metabolic disease. FEBS Lett 2008;582:2-9.
Tomas L, Edsfeldt A, Mollet IG, Perisic Matic L, Prehn C, Adamski J, Paulsson-Beme G, Hedin U, Nilsson J, Bengtsson E, Gonqalves I, Bjorkbacka H. Altered metabolism
distinguishes high-risk from stable carotid atherosclerotic plaques. Eur Heart J
2018;39:2301-10.
Teng F, Meng X, Kong L, Yu J. Progress and challenges of predictive biomarkers of anti PD- 1/PD-Ll immunotherapy: A systematic review. Cancer Lett 2018;414: 166-73.
Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N Engl J Med 2017;377:2500-1.
Zhu A, Lee D, Shim H. Metabolic positron emission tomography imaging in cancer detection and therapy response. Semin Oncol 2011 ;38 : 55— 69.

Claims

U30910WO Universitat Heidelberg Claims
1. Use of very long-chain fatty acid-containing lipids (VLCFA-containing lipids) as prognostic marker or predictive marker for a cancer therapy and/or for cancer therapy monitoring.
2. The use of claim 1, wherein said cancer therapy is an immuno-oncologic therapy, and/or wherein said use comprises response prediction to an immuno-oncologic therapy.
3. The use of claim 1 or 2, wherein cancer comprises locally advanced and/or metastatic cancer,
preferably renal cancer and cancer of the bladder, ureters, and renal pelvis.
4. The use of any of claims 1 to 3, wherein said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI), such as treatments targeting PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), CTLA4 (cytotoxic T- lymphocyte-associated protein 4), or LAG3 (lymphocyte-activation gene 3),
(a) as CPI monotherapy,
(b) as combinations of one CPI with a second CPI, and/or
(c) as combinations of one or more CPI with other targeted cancer therapeutics (e.g. kinase inhibitors, anti-VEGF antibodies, PPAR agonists) and/or
(d) as combinations of one or more CPI with chemotherapy.
5. The use of any of claims 1 to 4, comprising determining the metabolome in a sample of a mammalian subject,
wherein the sample preferably comprises blood, such as whole blood, plasma or serum; and/or wherein the mammalian subject is human.
6. The use of claim 5, wherein the metabolome is determined via mass spectrometry (MS), enzyme-linked immunosorbent assays (ELISA) or nuclear magnetic resonance (NMR) spectroscopy.
7. The use of any of the preceding claims, wherein the use is in combination with further markers,
preferably high PD-L1 expression, tumor mutational burden (TMB), microsatellite status, HLA (human leukocyte antigen) type, tumor gene expression signatures, and tumor infiltrating lymphocytes and/or combinations thereof.
8. An in vitro , in vivo or ex vivo method for the prognosis and/or therapy monitoring of cancer, comprising the following steps:
(a) providing a sample of a mammalian subject,
(b) determining the metabolome in said sample, and
(c) determining the amount of very long-chain fatty acid-containing lipids
(VLCFA-containing lipids) in the metabolome.
9. The method of claim 8, wherein said cancer therapy is an immuno-oncologic therapy, and/or wherein the method is preferably for the response prediction to an immuno-oncologic therapy, and/or wherein cancer comprises locally advanced and/or metastatic cancer,
preferably renal cancer and cancer of the bladder, ureters, and renal pelvis.
10. The method of claim 8 or 9, wherein in step (b) the metabolome is determined via mass spectrometry (MS), enzyme-linked immunosorbent assays (ELISA) or nuclear magnetic resonance (NMR) spectroscopy.
11. The method of any of claims 8 to 10, wherein in step (c) the amount of VLCFA- containing lipids in the metabolome is determined
via internal standard(s) (absolute quantification), or
relative to other measured metabolites in the same sample (relative quantification).
12. The method of any of claims 8 to 11, further comprising step (d):
(d) grouping the VLCFA-containing lipids determined in step (b) and/or (c) into clusters,
optionally further comprising step (e): (e) calculating the centroid of concentrations for the clusters.
13. The method of any of claims 8 to 12, wherein the sample comprises blood, such as whole blood, plasma or serum; or saliva,
and/or wherein the mammalian subject is human.
14. The method of any of claims 8 to 13, wherein said immuno-oncologic therapy comprises treatment with checkpoint inhibitors (CPI), such as treatments targeting PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), CTLA4 (cytotoxic T-lymphocyte-associated protein 4), or LAG3 (lymphocyte-activation gene 3),
(a) as CPI monotherapy,
(b) as combinations of one CPI with a second CPI, and/or
(c) as combinations of one or more CPI with other targeted cancer therapeutics (e.g. kinase inhibitors, anti-VEGF antibodies, PPAR agonists) and/or
(d) as combinations of one or more CPI with chemotherapy
15. The method of any of claims 8 to 14, comprising determining further markers, preferably high PD-L1 expression, tumor mutational burden (TMB), microsatellite status, HLA (human leukocyte antigen) type, tumor gene expression signatures, and tumor infiltrating lymphocytes and/or combinations thereof.
PCT/EP2020/056895 2019-03-14 2020-03-13 Vlcfa-containing lipids as prognostic and predictive marker for cancer therapies WO2020182995A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP19162764 2019-03-14
EP19162764.5 2019-03-14

Publications (1)

Publication Number Publication Date
WO2020182995A1 true WO2020182995A1 (en) 2020-09-17

Family

ID=65812169

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/056895 WO2020182995A1 (en) 2019-03-14 2020-03-13 Vlcfa-containing lipids as prognostic and predictive marker for cancer therapies

Country Status (1)

Country Link
WO (1) WO2020182995A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011098509A1 (en) * 2010-02-11 2011-08-18 Katholieke Universiteit Leuven Phospholipid profiling and cancer
WO2012038525A1 (en) * 2010-09-24 2012-03-29 Katholieke Universiteit Leuven Cancer phospholipidome
CN106802334A (en) * 2015-11-26 2017-06-06 中国医学科学院基础医学研究所 The individuation mark that one group of monitoring Cancerous disease progress and/or drug resistance are produced

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011098509A1 (en) * 2010-02-11 2011-08-18 Katholieke Universiteit Leuven Phospholipid profiling and cancer
WO2012038525A1 (en) * 2010-09-24 2012-03-29 Katholieke Universiteit Leuven Cancer phospholipidome
CN106802334A (en) * 2015-11-26 2017-06-06 中国医学科学院基础医学研究所 The individuation mark that one group of monitoring Cancerous disease progress and/or drug resistance are produced

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
BALAR AVCASTELLANO DO'DONNELL PHGRIVAS PVUKY JPOWLES TPLIMACK ERHAHN NMDE WIT RPANG L: "First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study", LANCET ONCOL, vol. 18, 2017, pages 1483 - 92, XP085258255, DOI: 10.1016/S1470-2045(17)30616-2
BETOF ASNIPP RDGIOBBIE-HURDER AJOHNPULLE RANRUBIN KRUBINSTEIN SMFLAHERTY KTLAWRENCE DPJOHNSON DBSULLIVAN RJ: "Impact of Age on Outcomes with Immunotherapy for Patients with Melanoma", ONCOLOGIST, vol. 22, 2017, pages 963 - 71
D. SEPAROVIC; J. BIELAWSKI; J. S. PIERCE; S. MERCHANT; A. L. TARCA; G. BHATTI; B. OGRETMEN; M. KORBELIK: "Enhanced tumor cures after Foscan photodynamic therapy combined with the ceramide analog LCL29. Evidence from mouse squamous cell carcinomas for sphingolipids as biomarkers of treatment response", INTERNATIONAL JOURNAL OF ONCOLOGY, vol. 38, no. 2, 6 December 2010 (2010-12-06), GR, pages 521 - 527, XP055259853, ISSN: 1019-6439, DOI: 10.3892/ijo.2010.863 *
FRITSCHE KL: "The science of fatty acids and inflammation", ADV NUTR, vol. 6, 2015, pages 293S - 301S, XP055583170, DOI: 10.3945/an.114.006940
GALON JCOSTES ASANCHEZ-CABO FKIRILOVSKY AMLECNIK BLAGORCE-PAGES CTOSOLINI MCAMUS MBERGER AWIND P: "Type, density, and location of immune cells within human colorectal tumors predict clinical outcome", SCIENCE, vol. 313, 2006, pages 1960 - 4
GIANNAKIS MLI HJIN CGOPAL SDESAI KHORAK CWIND-ROTOLO MVAN ALLEN EMCLISH CHODI FS: "Metabolomic correlates of response in nivolumab-treated renal cell carcinoma and melanoma patients", J CLIN ONCOL, vol. 35, 2017, pages 3036 - 6
HAAS RSMITH JROCHER-ROS VNADKARNI SMONTERO-MELENDEZ TD'ACQUISTO FBLAND EJBOMBARDIERI MPITZALIS CPERRETTI M: "Lactate Regulates Metabolic and Pro-inflammatory Circuits in Control of T Cell Migration and Effector Functions", P LOS BIOI, vol. 13, 2015, pages e 1 002202
HAKIMI AAREZNIK ELEE C-HCREIGHTON CJBRANNON ARLUNA AAKSOY BALIU EMSHEN RLEE W: "An Integrated Metabolic Atlas of Clear Cell Renal Cell Carcinoma", CANCER CELL, vol. 29, 2016, pages 104 - 16, XP029383984, DOI: 10.1016/j.ccell.2015.12.004
JAYASHREE BALASUBRAMANYAM ET AL: "Monitoring of changes in lipid profiles during PLK1 knockdown in cancer cells using DESI MS", ANALYTICAL AND BIOANALYTICAL CHEMISTRY, SPRINGER, DE, vol. 408, no. 20, 8 June 2016 (2016-06-08), pages 5623 - 5632, XP036000816, ISSN: 1618-2642, [retrieved on 20160608], DOI: 10.1007/S00216-016-9665-Y *
JOHNSON CHSPILKER MEGOETZ LPETERSON SNSIUZDAK G: "Metabolite and Microbiome Interplay in Cancer Immunotherapy", CANCER RES, vol. 76, 2016, pages 6146 - 52
JOO ERN ANG ET AL: "Modulation of Plasma Metabolite Biomarkers of the MAPK Pathway with MEK Inhibitor RO4987655: Pharmacodynamic and Predictive Potential in Metastatic Melanoma", MOLECULAR CANCER THERAPEUTICS, vol. 16, no. 10, 21 June 2017 (2017-06-21), US, pages 2315 - 2323, XP055611503, ISSN: 1535-7163, DOI: 10.1158/1535-7163.MCT-16-0881 *
LI BQIU BLEE DSMWALTON ZEOCHOCKI JDMATHEW LKMANCUSO AGADE TPFKEITH BNISSIM I: "Fructose-1,6-bisphosphatase opposes renal carcinoma progression", NATURE, vol. 513, 2014, pages 251 - 5
LIN LIN ET AL: "LC-MS based serum metabonomic analysis for renal cell carcinoma diagnosis, staging, and biomarker discovery", JOURNAL OF PROTEOME RESEARCH, vol. 10, no. 3, 4 March 2011 (2011-03-04), pages 1396 - 1405, XP055017488, ISSN: 1535-3893, DOI: 10.1021/pr101161u *
LYSSIOTIS CAKIMMELMAN AC: "Metabolic Interactions in the Tumor Microenvironment", TRENDS CELL BIOL, vol. 27, 2017, pages 863 - 75, XP085234112, DOI: 10.1016/j.tcb.2017.06.003
MAIA MCALMEIDA LBERGEROT PGDIZMAN NPAL SK: "Relationship of tumor mutational burden (TMB) to immunotherapy response in metastatic renal cell carcinoma (mRCC", J CLIN ONCOL, vol. 36, 2018, pages 662 - 2
NIZIOL JBONIFAY VOSSOLINSKI KOSSOLINSKI TOSSOLINSKA ASUNNER JBEECH IARENDOWSKI ARUMAN T: "Metabolomic study of human tissue and urine in clear cell renal carcinoma by LC-HRMS and PLS-DA", ANAL BIOANAL CHEM, vol. 410, 2018, pages 3859 - 69, XP055668109, DOI: 10.1007/s00216-018-1059-x
PARISI LRLI NATILLA-GOKCUMEN GE: "Very Long Chain Fatty Acids Are Functionally Involved in Necroptosis", CELL CHEM BIOL, vol. 24, 2017, pages 1445 - 8
PAVLOVA NNTHOMPSON CB: "The Emerging Hallmarks of Cancer Metabolism", CELL METAB, vol. 23, 2016, pages 27 - 47, XP029385101, DOI: 10.1016/j.cmet.2015.12.006
PINHEIRO J., NLME : LINEAR AND NONLINEAR MIXED EFFECTS MODELS, 2009, Retrieved from the Internet <URL:http://cranrprojectorglweblpackageslnlmelindexhtml>
SAHU DLOTAN YWITTMANN BNERI BHANSEL DE: "Metabolomics analysis reveals distinct profiles of nonmuscle-invasive and muscle-invasive bladder cancer", CANCER MED, vol. 6, 2017, pages 2106 - 20, XP055615023, DOI: 10.1002/cam4.1109
SAITO KOSUKE ET AL: "Lipid profiling of pre-treatment plasma reveals biomarker candidates associated with response rates and hand-foot skin reactions in sorafenib-treated patients", CANCER CHEMOTHERAPY AND PHARMACOLOGY, SPRINGER VERLAG, BERLIN, vol. 82, no. 4, 30 July 2018 (2018-07-30), pages 677 - 684, XP036589269, ISSN: 0344-5704, [retrieved on 20180730], DOI: 10.1007/S00280-018-3655-Z *
SCHMERLER DNEUGEBAUER SLUDEWIG KBREMER-STRECK SBRUNKHORST FMKIEHNTOPF M: "Targeted metabolomics for discrimination of systemic inflammatory disorders in critically ill patients", J LIPID RES, vol. 53, 2012, pages 1369 - 75
SONODA JPEI LEVANS RM: "Nuclear receptors: decoding metabolic disease", FEBS LETT, vol. 582, 2008, pages 2 - 9, XP022399002, DOI: 10.1016/j.febslet.2007.11.016
TENG FMENG XKONG LYU J.: "Progress and challenges of predictive biomarkers of anti PD-1/PD-L1 immunotherapy: A systematic review", CANCER LETT, vol. 414, 2018, pages 166 - 73, XP085320038, DOI: 10.1016/j.canlet.2017.11.014
TOMAS LEDSFELDT AMOLLET IGPERISIC MATIC LPREHN CADAMSKI JPAULSSON-BERNE GHEDIN UNILSSON JBENGTSSON E: "Altered metabolism distinguishes high-risk from stable carotid atherosclerotic plaques", EUR HEART J, vol. 39, 2018, pages 2301 - 10
YARCHOAN MHOPKINS AJAFFEE EM: "Tumor Mutational Burden and Response Rate to PD-1 Inhibition", NENGL J MED, vol. 377, 2017, pages 2500 - 1, XP055628897, DOI: 10.1056/NEJMc1713444
YASUYUKI KONDO ET AL: "Serum fatty acid profiling of colorectal cancer by gas chromatography/mass spectrometry", BIOMARKERS IN MEDICINE, vol. 5, no. 4, 1 August 2011 (2011-08-01), UK, pages 451 - 460, XP055614864, ISSN: 1752-0363, DOI: 10.2217/bmm.11.41 *
YUJI YAMASHITA ET AL: "Differences in elongation of very long chain fatty acids and fatty acid metabolism between triple-negative and hormone receptor-positive breast cancer", BMC CANCER, vol. 17, no. 1, 29 August 2017 (2017-08-29), XP055614842, DOI: 10.1186/s12885-017-3554-4 *
ZHU ALEE DSHIM H: "Metabolic positron emission tomography imaging in cancer detection and therapy response", SEMIN ONCOL, vol. 38, 2011, pages 55,69

Similar Documents

Publication Publication Date Title
Tumas et al. Metabolomics in pancreatic cancer biomarkers research
Chang et al. Secretome-based identification of ULBP2 as a novel serum marker for pancreatic cancer detection
Hudler et al. Proteomic approaches in biomarker discovery: new perspectives in cancer diagnostics
Bansal et al. Low-and high-grade bladder cancer determination via human serum-based metabolomics approach
Korse et al. Multicenter evaluation of a new progastrin-releasing peptide (ProGRP) immunoassay across Europe and China
AU2016214140B2 (en) Biomarkers for pancreatic cancer
Mock et al. Serum very long-chain fatty acid-containing lipids predict response to immune checkpoint inhibitors in urological cancers
JP2020510816A (en) Methods for detection and treatment of lung cancer
Roe et al. Mesothelin-related predictive and prognostic factors in malignant mesothelioma: a nested case–control study
CN110007083B (en) Hyaluronic acid as biomarker for metastatic breast cancer
Hao et al. IPO-38 is identified as a novel serum biomarker of gastric cancer based on clinical proteomics technology
Li et al. Liquid biopsy at the frontier in renal cell carcinoma: recent analysis of techniques and clinical application
Jin et al. A novel panel of biomarkers in distinction of small well-differentiated HCC from dysplastic nodules and outcome values
Bryan The current state of clinical application of serum biomarkers for canine lymphoma
Shimura et al. Serum transthyretin level is associated with prognosis of patients with gastric cancer
Duangkumpha et al. Discovery and qualification of serum protein biomarker candidates for cholangiocarcinoma diagnosis
Shim et al. Half-life of serum alpha-fetoprotein: an early prognostic index of recurrence and survival after hepatic resection for hepatocellular carcinoma
Li et al. High number of circulating tumor cells predicts poor survival of cutaneous melanoma patients in China
Hashimoto et al. Use of squamous cell carcinoma antigen as a biomarker of chemotherapy response in patients with metastatic cervical carcinoma
Ma et al. Elevated soluble programmed death-ligand 1 levels indicate immunosuppression and poor prognosis in hepatocellular carcinoma patients undergoing transcatheter arterial chemoembolization
Tang et al. Immunometabolic coevolution defines unique microenvironmental niches in ccRCC
EP2904397B1 (en) Means and methods for diagnosing recurrence of prostate cancer after prostatectomy
Chen et al. The expression and prognostic value of REXO4 in hepatocellular carcinoma
WO2020182995A1 (en) Vlcfa-containing lipids as prognostic and predictive marker for cancer therapies
WO2019115679A1 (en) A signature to assess prognosis and therapeutic regimen in liver cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20711875

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20711875

Country of ref document: EP

Kind code of ref document: A1