WO2020104777A1 - Compounds for telomere length-related treatment of cancer - Google Patents

Compounds for telomere length-related treatment of cancer

Info

Publication number
WO2020104777A1
WO2020104777A1 PCT/GB2019/053261 GB2019053261W WO2020104777A1 WO 2020104777 A1 WO2020104777 A1 WO 2020104777A1 GB 2019053261 W GB2019053261 W GB 2019053261W WO 2020104777 A1 WO2020104777 A1 WO 2020104777A1
Authority
WO
WIPO (PCT)
Prior art keywords
ddri
telomere length
use according
chromosome
group
Prior art date
Application number
PCT/GB2019/053261
Other languages
French (fr)
Inventor
Christopher PEPPER
Duncan Martin Baird
Christopher Fegan
Joseph Thomas BIRKETT
Original Assignee
Telonostix Ltd
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 Telonostix Ltd filed Critical Telonostix Ltd
Publication of WO2020104777A1 publication Critical patent/WO2020104777A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the invention relates to a DNA damage response inhibitor (DDRi) for use, optionally in combination with a DNA damaging agent and/or a DNA damaging procedure, in treating cancer in a patient, characterized in that the patient’s cancerous cells have a mean telomere length, determined using high- resolution telomere length analysis, that is less than or equal to a threshold value; and to methods for treating cancer in said patient by administering a DDRi, optionally in combination with a DNA damaging agent and/or a DNA damaging procedure.
  • DDRi DNA damage response inhibitor
  • Telomeres are nucleoprotein structures composed of repetitive DNA sequences that cap the ends of linear eukaryotic chromosomes, protecting them from deterioration or fusion with adjacent chromosomes.
  • the repeat sequence of nucleotides in telomeres is TTAGGG, the complementary DNA strand being CCCTAA, with a single-stranded CCCTAA overhang. This sequence of CCCTAA is repeated approximately 2,500 times in humans.
  • the average telomere length declines from about 1 1 kilobases at birth to less than 4 kilobases in old age, with the average rate of decline being greater in men than in women.
  • telomere ends are vulnerable to degradation; telomeres however prevent this happening by themselves being degraded during each stage of cell division, essentially ‘capping’ the chromosome.
  • Telomere ends are, however, maintained in certain cell types such as germ cells, stem cells and certain white blood cells, by the reverse transcriptase telomerase that catalyses the RNA templated addition of telomere repeats.
  • Telomere length is a key determinant of telomeric function and it has been shown that short dysfunctional telomeres can drive genomic instability and tumourigenesis in mouse models. Furthermore, deregulation of telomerase has been shown to drive oncogenesis.
  • telomere loss in somatic cells has been linked to replicative senescence preventing genomic instability and cancer. Conversely, it has also been shown that malignant cells can by-pass this senescence and become immortalised via telomere extension using aberrant activation of telomerase.
  • telomere length can provide prognostic information in many human malignancies including Chronic lymphocytic leukaemia (CLL) 1-8
  • CLL chronic lymphocytic leukaemia
  • CLL has a heterogeneous clinical course with survival ranging from a few months to many decades.
  • Treatment strategies vary with staging or disease progression and include chemotherapy, radiotherapy, monoclonal antibody therapy or bone marrow transplantation, although, early stage patients often receive no treatment.
  • telomere length analysis allows complete resolution of telomere lengths at specific chromosome ends, including telomeres in a length range, termed the fusogenic range, at which telomere end-end fusions occur 9 11 .
  • STELA therefore permits detection of shortened telomeres that are potentially dysfunctional and capable of fusion.
  • telomere length and fusion analysis have used telomere length and fusion analysis to identify the longest mean telomere length at which telomere end-end fusion events can be detected for a number of chromosomes (examples are shown in Table 1 ). Further we have shown that CLL patients with telomere lengths within this fusogenic range (i.e. less than or equal to the to the longest mean telomere length at which telomere end-end fusion events can be detected), have inferior clinical outcomes when compared to patients with telomere lengths above the longest mean telomere length at which telomere end-end fusion events can be detected 13 . Indeed, these investigations demonstrated that telomere lengths can accurately predict response to chemotherapy 14 and chemo-immunotherapy 15 , with short telomere patients having significantly worse progression-free survival and overall survival.
  • telomere length profiles within the fusogenic range could particularly benefit from the administration of a DDRi, optionally when used in combination with a DNA damaging agent (e.g. chemotherapy agent) and/or a DNA damaging procedure (e.g. radiotherapy).
  • a DNA damaging agent e.g. chemotherapy agent
  • a DNA damaging procedure e.g. radiotherapy
  • cells with short telomeres are shown to be inherently more sensitive to DDRi and also the combination of a DDRi and a DNA damaging agent than cells with telomeres above the fusogenic range.
  • a DNA damage response inhibitor for use in treating cancer in a patient whose cancer cells have a mean telomere length, determined using high-resolution telomere length analysis, that is less than or equal to a threshold value selected according to the chromosome to be tested from the group consisting of: i) 3.81 kb, preferably 2.26 kb, for XpYp;
  • telomere length threshold determined using any two or more of the above threshold values identified in i) to vi) depending upon the combination of different chromosomes that are tested.
  • the threshold values indicated in i) to vi) above each represents an established chromosome-specific upper limit of the threshold value. Therefore, if the patient’s mean telomere length is determined using a mean average value derived from two or more of the above chromosomes, then the mean telomere length threshold is calculated as the mean of the relevant chromosome- specific upper limits identified in i) to v) above. For example:
  • telomere length threshold is calculated for XpYp, 17p, 2p and 16p
  • telomere end-end fusion events represent the longest mean telomere length at which telomere end-end fusion events can be detected for a specific chromosome.
  • a range termed herein a fusogenic range, in which shortened telomeres become dysfunctional.
  • the above said threshold value is determined for any other chromosome (such as one or more of 1 1 q, 9p, 5p and 12q) and once this value is established the above invention is worked by using a DNA damage response inhibitor (DDRi) for use in treating cancer in a patient whose cancer cells have a mean telomere length, determined using high-resolution telomere length analysis, that is less than or equal to the said threshold value established for said any other chromosome or a mean telomere length threshold determined using any two or more of the above threshold values identified in i) to vii) depending upon the combination of different chromosomes that are tested.
  • DDRi DNA damage response inhibitor
  • telomere length analysis is known to be suitable for determining mean telomere length for most, if not all, chromosomes, in particular XpYp, 17p, 2p, 16p, 18q, 7q, 1 1 q, 9p, 5p and 12q.
  • the invention may be practiced by those skilled in the art by analyzing any other chromosome to find the relevant threshold or combination of chromosomes (optionally together with one or more of XpYp, 17p, 2p, 16p, 18q and/or 7q) to find the mean threshold: the threshold value being the longest mean telomere length at which telomere end-end fusions can be detected in samples of tissue from a plurality of individuals presenting with the same cancer condition.
  • the cancer is a B-cell malignant condition
  • a condition includes, but is not limited to, diffuse large B-cell lymphoma (DLBCL), follicular lymphoma, marginal zone B- cell lymphoma (MZL), mucosa-associated lymphatic tissue lymphoma (MALT), chronic lymphocytic leukaemia, (CLL), Mantle cell lymphoma (MCL), B cell Acute lymphoblastic leukaemia (B-ALL), Non- Hodgkin’s Lymphoma (NHL), Hodgkin’s Lymphoma (HL) and Multiple myeloma (MM).
  • DLBCL diffuse large B-cell lymphoma
  • MZL marginal zone B- cell lymphoma
  • MALT mucosa-associated lymphatic tissue lymphoma
  • CLL chronic lymphocytic leukaemia
  • MCL Mantle cell lymphoma
  • B-ALL B cell Acute lymphoblastic leukaemia
  • NHL Ho
  • the B-cell malignant condition is CLL, the cancerous cells of which may or may not possess large scale chromosomal abnormalities / deletions.
  • Chromosomal abnormalities that are often associated with CLL include, but are not limited to, deletions of 1 1 q and 17p chromosomes.
  • the DDRi may be any compound or complex that has an inhibitory effect on one or more cellular DNA damage response pathways (e.g. signaling or repair).
  • the DDRi is selected from the group consisting of inhibitors of one or more of the following proteins: ATM serine/threonine kinase (ATM), ATR serine/threonine-protein kinase (ATR), Checkpoint kinase 1 (Chk1 ), Checkpoint kinase 2 (CHk2), DNA-dependent protein kinase catalytic subunit (DNA-PKcs), DNA polymerase theta (POLQ), Wee1 -like protein kinase (WEE1A, also known as WEE1 ), Wee1 -like protein kinase 2 (WEE1 B, also known as WEE2), and poly ADP ribose polymerase (PARP). More preferably, the DDRi is selected from the group consisting of inhibitors of one or more of Chk
  • the Chk1 inhibitor is GDC-0575 (i.e. (R)-N-(4-(3-aminopiperidin-1 - yl)-5-bromo-1 H-pyrrolo[2,3-b]pyridin-3-yl)cyclopropanecarboxamide (CAS No. 1 196541 -47-5)).
  • GDC-0575 i.e. (R)-N-(4-(3-aminopiperidin-1 - yl)-5-bromo-1 H-pyrrolo[2,3-b]pyridin-3-yl)cyclopropanecarboxamide (CAS No. 1 196541 -47-5).
  • the chemical structure of GDC-0575 is shown below:
  • the WEE1A inhibitor is MKK-1775 (i.e. 1 -[6-(2-Hydroxypropan-2- yl)pyridin-2-yl]-6-[4-(4-methylpiperazin-1 -yl)anilino]-2-prop-2-enylpyrazolo [3,4-d]pyrimidin-3-one (CAS No. 955385-80-7)), which is commonly known as
  • Adavosertib The chemical structure of MKK-1775 is shown below.
  • Preferred PARP inhibitors are selected from the group consisting of: olaparib, rucaparib, niraparib, talazoparib, veliparib, 3-aminobenzamide, and iniparib.
  • the PARP inhibitor is olaparib.
  • the chemical structures of each of the preferred PARP inhibitors are shown below:
  • the DDRi is used in combination with a DNA damaging agent and/or a DNA damaging procedure.
  • a DNA damaging procedure is radiotherapy.
  • the DDRi is used in combination with a DNA damaging agent.
  • Preferred DNA damaging agents are selected from the group consisting of nucleoside analogues and alkylating agents.
  • a particularly preferred nucleoside analogue is fludarabine.
  • Particularly preferred alkylating agents are selected from the group consisting of chlorambucil, bendamustine, mafosfamide, cyclophosphamide and 4-hydroxy-cyclophosphamide (i.e. the active metabolite of both mafosfamide and, following hepatic activation, cyclophosphamide).
  • telomere length analysis is typically measured using a single telomere length analysis (STELA) method, which allows complete resolution of telomere lengths at specific chromosome ends 9 11 .
  • STELA single telomere length analysis
  • any other method that can measure the full range of telomere length from one telomere repeat to several kb of telomere length may be utilised.
  • telomere length is detected for a single chromosome.
  • a mean telomere length may be detected for a plurality of different chromosomes and, in such embodiments, an average mean telomere length is calculated.
  • mean telomere length is determined for the XpYp chromosome, either alone or in combination with one or more further chromosomes. In preferred embodiments, the mean telomere length is determined using at least one, any two, any three, any four, any five, any six, any seven, any eight, any nine or all ten of the following chromosomes: XpYp, 17p, 2p, 16p, 18q, 7q, 1 1 q, 9p, 5p and 12q.
  • the mean telomere length is determined using at least any one, any two, any three, any four, five or all six of the following chromosomes: XpYp, 17p, 2p, 16p, 18q and 7q chromosomes.
  • telomere length Whilst in principle telomere length can be assessed using any type of cell, the mean telomere length will typically be determined from at least a sample of the cancerous cell type. Therefore, in preferred embodiments, the mean telomere length is determined using at least a sample of B-cells isolated from the patient to be treated.
  • a method for treating cancer in a patient comprising: a. using high-resolution telomere length analysis to determine the mean telomere length (in kb) of at one chromosome selected from XpYp, 17p, 2p, 16p, 18q and 7q in at least a sample of cancer cells from said patient; and
  • telomere length is less than or equal to a threshold value selected according to the chromosome that was tested from the group consisting of:
  • telomere length threshold determined using any two or more of the above threshold values identified in i) to vi) depending upon the combination of different chromosomes that are tested
  • the cancer may be any type of cancer but according to a preferred embodiment, the form of cancer is a 13- cell malignant condition.
  • the B-cell malignant condition is CLL, the cancerous cells of which may or may not possess large scale chromosomal abnormalities / deletions, such abnormalities / deletions usually being found in, but not limited to, chromosomes 1 1 q and 17p.
  • the DDRi is selected from the group consisting of inhibitors of one or more proteins selected from the group consisting of an ATM, ATR, Chk1 , CHk2, DNA-PKcs, POLQ, WEE1A, WEE1 B, and PARP. More preferably the DDRi is selected from the group consisting of inhibitors of one or more of Chk1 , WEE1A and PARP. In a particularly preferred embodiment, the DDRi is a PARP inhibitor.
  • a preferred Chk1 inhibitor is GDC-0575.
  • a preferred WEE1A inhibitor is MKK- 1775.
  • Preferred PARP inhibitors are selected from the group consisting olaparib, rucaparib, niraparib, talazoparib, veliparib, 3-aminobenzamide, and iniparib.
  • the PARP inhibitor is olaparib.
  • the DDRi is administered in combination with a DNA damaging agent and/or before, during or after a DNA damaging procedure.
  • a DNA damaging procedure is radiotherapy.
  • the DDRi is administered in combination with a DNA damaging agent.
  • preferred DNA damaging agents are selected from the group consisting of nucleoside analogues and alkylating agents.
  • a particularly preferred nucleoside analogue is fludarabine.
  • Particularly preferred alkylating agents are selected from the group consisting of chlorambucil, bendamustine, mafosfamide, cyclophosphamide and 4-hydroxy-cyclophosphamide.
  • telomere length is detected for a single chromosome.
  • a mean telomere length may be detected for a plurality of different chromosomes and, in such embodiments, an average mean telomere length is calculated.
  • mean telomere length is determined for the XpYp chromosome, either alone or in combination with one or more of said further chromosomes.
  • the mean telomere length is determined using at least one, two, three, four, five, six, seven, eight, nine or all ten of the following chromosomes: XpYp, 17p, 2p, 16p, 18q, 7q, 1 1 q, 9p, 5p and 12q. In more preferred embodiments, the mean telomere length is determined using at least one, two, three, four, five or all six of the following chromosomes: XpYp, 17p, 2p, 16p, 18q and 17q chromosomes.
  • Mean telomere length is typically determined from at least a sample of cancer cell type. Therefore, in preferred embodiments, the mean telomere length is determined using at least a sample of B-cells isolated from the patient to be treated.
  • any feature disclosed herein may be replaced by an alternative feature serving the same or a similar purpose.
  • Figure 1 Comparative cytotoxicity of (A) fludarabine (B) chlorambucil (C) bendamustine (D) mafosfamide (E) rituximab and (F) olaparib in monoculture and co-culture for 48h.
  • FIG. 1 Comparison of the cytotoxic effect in primary CLL cell monocultures treated with (A) fludarabine (B) chlorambucil (C) bendamustine (D) mafosfamide (E) olaparib in patient samples grouped according to telomere length.
  • FIG. 3 Comparison of the cytotoxic effect in primary CLL cells co-cultured on CD40L-expressing mouse fibroblasts and treated with (A) fludarabine (B) chlorambucil (C) bendamustine (D) mafosfamide (E) olaparib in patient samples grouped according to telomere length.
  • FIG. 4 Comparison of the synergistic cytotoxic effect in primary CLL cell monocultures treated with a combination of olaparib and (A) fludarabine, (B) chlorambucil, (C) bendamustine, and (D) mafosfamide.
  • A fludarabine
  • B chlorambucil
  • C bendamustine
  • D mafosfamide.
  • the short telomere subset showed significantly enhanced synergy with all four drug combinations in comparison with that observed for the long telomere subset.
  • FIG. 5 Comparison of the synergistic cytotoxic effect in primary CLL cells co-cultured on CD40L-expressing mouse fibroblasts that were treated with a combination of olaparib and (A) fludarabine, (B) chlorambucil, (C) bendamustine, and (D) mafosfamide.
  • the short telomere subset showed strong synergy with all four drug combinations whereas the long telomere subset demonstrated additive effects or weak synergy.
  • Figure 6 Comparison of the synergistic cytotoxic effect in primary CLL cell monocultures treated with a combination of MK-1775 and (A) bendamustine or (B) mafosfamide. The short telomere subset showed significantly enhanced synergy with both drug combinations in comparison with that observed for the long telomere subset.
  • FIG. 7 Comparison of the synergistic cytotoxic effect in primary CLL cell monocultures treated with a combination of GDC-0575 and (A) bendamustine or (B) mafosfamide.
  • the short telomere subset showed significantly enhanced synergy with both drug combinations in comparison with that observed for the long telomere subset.
  • CLL Peripheral blood samples from CLL consenting patients, in accordance with the Declaration of Helsinki and as approved by the South East Wales local research ethics committee (LREC# 02/4806).
  • CLL was defined by clinical criteria as well as cellular morphology, and also the co-expression of CD19 and CD5 in lymphocytes simultaneously displaying restriction of light-chain rearrangement. All of the samples were collected at, or close to, the time of diagnosis and staging was based on the Binet classification system 16 .
  • PBMCs Peripheral blood mononuclear cells
  • CD19 + B-cells were positively selected by using CD19 + dynabeads from Life Technologies. 10pL of the desired bead was used for the isolation of 500,000 target cells form isolated PBMCs. The cell preparation containing the cells of interest and the washed dynabeads was incubated for 20 minutes at 4°C. Cells were isolated by placing the microtube back into the magnetic particle concentrator for 2 minutes. After this time, the supernatant was aspirated and discarded and the microtube was removed from the magnetic particle concentrator.
  • telomere length analysis at the XpYp telomere we used an adaptation of the chromosome- specific single telomere length analysis (STELA) assay, as previously described 9 11 , to allow for high-throughput analysis (HT-STELA).
  • STELA protocol was adapted to use telomere-adjacent primers specific for the XpYp telomere (XpYpC: 5' -C AG G G AC C G G GAC AAAT AG AC-3' ) , in triplicate 30pl PCR reactions each containing 30 ng of genomic DNA.
  • Thermal cycling conditions were: 23 cycles of 94°C for 20 s, 65°C for 30 s and 68°C for 5 mins. Amplified fragments were resolved using capillary gel electrophoresis and mean telomere length determined using PROSize software (AATI, Ankeny, Iowa, USA).
  • cytotoxicity of all the agents were first tested in the following monoculture conditions: 1x10 6 CLL cells were resuspended in 1 mL of cell culture medium - RPMI (1640 Sigma), which was supplemented with: 2mM L-Glutamine (Life Technologies); 100units/ml Penicillin and 100g/mL streptomycin (Life Technologies); and 10% foetal Calf Serum (FCS) (Life Technologies) supplemented with 5ng/pL IL-4 (Biosource).
  • FCS foetal Calf Serum
  • CD40L-expressing mouse fibroblast cells were suspended in 5m L of fibroblast media and irradiated (8000RADs). 1x10 5 irradiated cells were then seeded into a 24-well plate. The plate was incubated overnight at 37°C with 5% CO2 to allow the fibroblast cells to adhere to the well surface.
  • the in vitro cytotoxicity of a range of DNA damaging agents was assessed in monoculture (i.e. CLL cells alone) and co-culture (i.e. CLL cells on CD40L- expressing mouse fibroblasts) to determine whether sensitivity to these agents was related to the telomere length profiles of the tumour cells. Subsequently, these agents were combined with various DDRi compounds, in particular olaparib (a PARP inhibitor), GDC-0575 (a Chk1 inhibitor) and MKK-1775 (a WEE1A inhibitor), in the same in vitro models. Analysis of the efficacy of chemotherapy agents, rituximab and the PARP inhibitor, olaparib, to kill primary CLL cells in monoculture and CD40L- expressing co-culture
  • olaparib was shown to enhance the cytotoxic effect when applied to CLL cells in co-culture. Although not bound to any particular theory (at least because the mechanism for this increased potency was not explored in this study), one may speculate that this increased cellular activation and tumour cell division induced by the co-culture conditions may make cells more prone to DNA damage and hence more reliant on DNA repair mechanisms. Therefore, DDRis such as PARP inhibitors may, even as monotherapies, be more effective in proliferating cells within a tumour.
  • telomere length The impact of telomere length on the ability of chemotherapy agents, rituximab and olaparib to kill primary CLL cells in monoculture and coculture
  • telomere length distribution in our CLL patient samples was analysed using single telomere length analysis (STELA) at the XpYp telomere, and the patients were divided into two groups based on telomere length (i.e. telomere length below 3.81 kb or a telomere length at/above 3.81 kb, 3.81 kb representing the upper threshold at which telomere end-end fusion events can be detected) before the response to drugs in our in vitro models was compared between said groups ( Figure 2 and Figure 3).
  • STELA single telomere length analysis
  • telomere short telomere group was significantly resistant to killing with the purine nucleoside analogue (fludarabine) and the three alkylating agents (chlorambucil, bendamustine and mafosfamide) in both monoculture ( Figure 2A-2D) and co-culture (Figure 3A-3D).
  • the same cells showed increased sensitivity to olaparib under both sets of in vitro conditions ( Figure 2E and Figure 3E respectively).
  • CLL cells derived from patients harbouring an 1 1 q deletion or a 17p deletion showed no significant difference in response to the drugs when the samples were grouped according to telomere length. This implies that CLL cells with short telomeres are inherently more resistant to the cytotoxic effect of DNA damaging agents, and more sensitive to the cytotoxic effects of DDRis such as PARP inhibitors, regardless of their cytogenetic background.
  • cytotoxicity efficacy was also evaluated in monoculture experiments using combinations comprising the WEE1A inhibitor MK-1775 ( Figure 6) or the Chk1 inhibitor GDC-0575 ( Figure 7) using the following DNA Damaging Agent: DDRi molar ratios:
  • DDRis such as PARP inhibitors, WEE1A inhibitors or Chk1 inhibitors, alone or in combination with DNA damaging agents/procedures, for the treatment of cancer.
  • telomeres are associated with genomic complexity in a number of human cancers. Therefore, one can speculate that patients with these adverse clinical characteristics may preferentially benefit from the combination of DNA damage response pathway inhibitors and DNA damaging agents.
  • tumour cell telomere length potentially could be used to risk- stratify patients (routinely or in clinical trials) and assign them to drug treatment regimens specifically designed to exploit their telomere length-associated weakness.
  • telomeres are associated with genetic complexity, high risk genomic aberrations, and short survival in chronic lymphocytic leukemia. Blood 2008; 1 11 :2246-52.
  • Telomere length predicts progression and overall survival in chronic lymphocytic leukemia: data from the UK LRF CLL4 trial. Leukemia. 2015 Dec; 29(12):241 1 -2414.

Abstract

The invention relates to a DNA damage response inhibitor (DDRi) for use, optionally in combination with a DNA damaging agent and/or a DNA damaging procedure, in treating cancer in a patient, characterized in that the patient's cancerous cells have a mean telomere length, determined using high- resolution telomere length analysis, that is less than or equal to a threshold value; and to methods for treating cancer in said patient by administering a DDRi and, optionally in combination with a DNA damaging agent and/or a DNA damaging procedure.

Description

Compounds for Telomere Length-Related Treatment of Cancer
The invention relates to a DNA damage response inhibitor (DDRi) for use, optionally in combination with a DNA damaging agent and/or a DNA damaging procedure, in treating cancer in a patient, characterized in that the patient’s cancerous cells have a mean telomere length, determined using high- resolution telomere length analysis, that is less than or equal to a threshold value; and to methods for treating cancer in said patient by administering a DDRi, optionally in combination with a DNA damaging agent and/or a DNA damaging procedure.
Background of Invention
Telomeres are nucleoprotein structures composed of repetitive DNA sequences that cap the ends of linear eukaryotic chromosomes, protecting them from deterioration or fusion with adjacent chromosomes. For humans, the repeat sequence of nucleotides in telomeres is TTAGGG, the complementary DNA strand being CCCTAA, with a single-stranded CCCTAA overhang. This sequence of CCCTAA is repeated approximately 2,500 times in humans. In humans, the average telomere length declines from about 1 1 kilobases at birth to less than 4 kilobases in old age, with the average rate of decline being greater in men than in women.
During replication of DNA, the ends of chromosomes are vulnerable to degradation; telomeres however prevent this happening by themselves being degraded during each stage of cell division, essentially ‘capping’ the chromosome. Telomere ends are, however, maintained in certain cell types such as germ cells, stem cells and certain white blood cells, by the reverse transcriptase telomerase that catalyses the RNA templated addition of telomere repeats. Telomere length is a key determinant of telomeric function and it has been shown that short dysfunctional telomeres can drive genomic instability and tumourigenesis in mouse models. Furthermore, deregulation of telomerase has been shown to drive oncogenesis. Additionally, the loss of telomeres in somatic cells has been linked to replicative senescence preventing genomic instability and cancer. Conversely, it has also been shown that malignant cells can by-pass this senescence and become immortalised via telomere extension using aberrant activation of telomerase.
Consistent with the role of telomere biology in tumour progression, there is now a substantial body of evidence indicating that telomere length can provide prognostic information in many human malignancies including Chronic lymphocytic leukaemia (CLL)1-8 CLL is the most common adult leukaemia, characterised by an accumulation of immuno-incompetent, monoclonal CD5+ B-lymphocytes. CLL has a heterogeneous clinical course with survival ranging from a few months to many decades. Treatment strategies vary with staging or disease progression and include chemotherapy, radiotherapy, monoclonal antibody therapy or bone marrow transplantation, although, early stage patients often receive no treatment. Early clinical intervention is required for patients with an aggressive form of the disease, whereas patients with a more benign form simply need monitoring for disease progression until a point is reached where appropriate treatment is administered. In this latter respect, it has been shown that early stage CLL intervention does not improve survival rate. It is therefore inappropriate to expose someone presenting with a disease that is unlikely to be life-threatening for up to 30 years with highly dangerous chemotherapeutic drugs.
In previous studies, we have developed single-molecule technologies that allow us to accurately detect the presence of critically shortened telomeres9 10 and to characterise telomere end-end fusions11 12 Single telomere length analysis (STELA) allows complete resolution of telomere lengths at specific chromosome ends, including telomeres in a length range, termed the fusogenic range, at which telomere end-end fusions occur9 11. STELA therefore permits detection of shortened telomeres that are potentially dysfunctional and capable of fusion. In particular, our previous investigations have used telomere length and fusion analysis to identify the longest mean telomere length at which telomere end-end fusion events can be detected for a number of chromosomes (examples are shown in Table 1 ). Further we have shown that CLL patients with telomere lengths within this fusogenic range (i.e. less than or equal to the to the longest mean telomere length at which telomere end-end fusion events can be detected), have inferior clinical outcomes when compared to patients with telomere lengths above the longest mean telomere length at which telomere end-end fusion events can be detected13. Indeed, these investigations demonstrated that telomere lengths can accurately predict response to chemotherapy14 and chemo-immunotherapy15, with short telomere patients having significantly worse progression-free survival and overall survival.
In the present investigations, we show that patients with short telomeres, i.e. telomere length profiles within the fusogenic range, could particularly benefit from the administration of a DDRi, optionally when used in combination with a DNA damaging agent (e.g. chemotherapy agent) and/or a DNA damaging procedure (e.g. radiotherapy). In particular, cells with short telomeres are shown to be inherently more sensitive to DDRi and also the combination of a DDRi and a DNA damaging agent than cells with telomeres above the fusogenic range.
Statements of Invention
According to a first aspect of the invention, there is therefore provided a DNA damage response inhibitor (DDRi) for use in treating cancer in a patient whose cancer cells have a mean telomere length, determined using high-resolution telomere length analysis, that is less than or equal to a threshold value selected according to the chromosome to be tested from the group consisting of: i) 3.81 kb, preferably 2.26 kb, for XpYp;
ii) 4.81 kb, preferably 2.57 kb, for 17p;
iii) 5.01 kb, preferably 3.01 kb, for 2p;
iv) 4.49 kb, preferably 2.94 kb, for 16p;
v) 4.47 kb, preferably 2.66 kb, for 18q;
vi) 4.27 kb, preferably 3.05 kb, for 7q; or
vii) a mean telomere length threshold determined using any two or more of the above threshold values identified in i) to vi) depending upon the combination of different chromosomes that are tested.
In particular, it has been found that patients with cancerous cells having a mean telomer length below a threshold value show a preferential response to treatment with said DDRi, optionally in combination with said DNA damaging agent and/or a DNA damaging procedure.
The threshold values indicated in i) to vi) above each represents an established chromosome-specific upper limit of the threshold value. Therefore, if the patient’s mean telomere length is determined using a mean average value derived from two or more of the above chromosomes, then the mean telomere length threshold is calculated as the mean of the relevant chromosome- specific upper limits identified in i) to v) above. For example:
• if the patient’s mean telomere length threshold is calculated for XpYp and 17p, the mean telomere length threshold is (3.81 +4.81 ) 1 2 = 4.31 kb;
• if the patient’s mean telomere length threshold is calculated for XpYp, 17p and 2p, the mean telomere length threshold is (3.81 +4.81 +5.01 ) / 3 = 4.54 kb;
• if the patient’s mean telomere length threshold is calculated for XpYp, 17p, 2p and 16p, the mean telomere length threshold is (3.81 +4.81 +5.01 +4.49) / 4 = 4.53 kb; and • if the patient’s mean telomere length threshold is calculated for XpYp, 17p, 2p, 16p and 18q, the mean telomere length threshold is (3.81 +4.81 +5.01 +4.49+4.47) / 5 = 4.52 kb.
These thresholds represent the longest mean telomere length at which telomere end-end fusion events can be detected for a specific chromosome. Below each threshold is a range, termed herein a fusogenic range, in which shortened telomeres become dysfunctional.
Additionally, or alternatively, the above said threshold value is determined for any other chromosome (such as one or more of 1 1 q, 9p, 5p and 12q) and once this value is established the above invention is worked by using a DNA damage response inhibitor (DDRi) for use in treating cancer in a patient whose cancer cells have a mean telomere length, determined using high-resolution telomere length analysis, that is less than or equal to the said threshold value established for said any other chromosome or a mean telomere length threshold determined using any two or more of the above threshold values identified in i) to vii) depending upon the combination of different chromosomes that are tested.
High-resolution telomere length analysis (STELA) is known to be suitable for determining mean telomere length for most, if not all, chromosomes, in particular XpYp, 17p, 2p, 16p, 18q, 7q, 1 1 q, 9p, 5p and 12q. Therefore, the invention may be practiced by those skilled in the art by analyzing any other chromosome to find the relevant threshold or combination of chromosomes (optionally together with one or more of XpYp, 17p, 2p, 16p, 18q and/or 7q) to find the mean threshold: the threshold value being the longest mean telomere length at which telomere end-end fusions can be detected in samples of tissue from a plurality of individuals presenting with the same cancer condition.
Whilst when working the invention the form of cancer to be treated is not limited to any particular cancer type, but in a preferred embodiment, the cancer is a B-cell malignant condition, such a condition includes, but is not limited to, diffuse large B-cell lymphoma (DLBCL), follicular lymphoma, marginal zone B- cell lymphoma (MZL), mucosa-associated lymphatic tissue lymphoma (MALT), chronic lymphocytic leukaemia, (CLL), Mantle cell lymphoma (MCL), B cell Acute lymphoblastic leukaemia (B-ALL), Non- Hodgkin’s Lymphoma (NHL), Hodgkin’s Lymphoma (HL) and Multiple myeloma (MM).
In a particularly preferred embodiment, the B-cell malignant condition is CLL, the cancerous cells of which may or may not possess large scale chromosomal abnormalities / deletions. Chromosomal abnormalities that are often associated with CLL include, but are not limited to, deletions of 1 1 q and 17p chromosomes.
The DDRi may be any compound or complex that has an inhibitory effect on one or more cellular DNA damage response pathways (e.g. signaling or repair). In a preferred embodiment, the DDRi is selected from the group consisting of inhibitors of one or more of the following proteins: ATM serine/threonine kinase (ATM), ATR serine/threonine-protein kinase (ATR), Checkpoint kinase 1 (Chk1 ), Checkpoint kinase 2 (CHk2), DNA-dependent protein kinase catalytic subunit (DNA-PKcs), DNA polymerase theta (POLQ), Wee1 -like protein kinase (WEE1A, also known as WEE1 ), Wee1 -like protein kinase 2 (WEE1 B, also known as WEE2), and poly ADP ribose polymerase (PARP). More preferably, the DDRi is selected from the group consisting of inhibitors of one or more of Chk1 , WEE1A and PARP. In a particularly preferred embodiment, the DDRi is a PARP inhibitor.
Preferably, the Chk1 inhibitor is GDC-0575 (i.e. (R)-N-(4-(3-aminopiperidin-1 - yl)-5-bromo-1 H-pyrrolo[2,3-b]pyridin-3-yl)cyclopropanecarboxamide (CAS No. 1 196541 -47-5)). The chemical structure of GDC-0575 is shown below:
Figure imgf000008_0001
Preferably the WEE1A inhibitor is MKK-1775 (i.e. 1 -[6-(2-Hydroxypropan-2- yl)pyridin-2-yl]-6-[4-(4-methylpiperazin-1 -yl)anilino]-2-prop-2-enylpyrazolo [3,4-d]pyrimidin-3-one (CAS No. 955385-80-7)), which is commonly known as
Adavosertib. The chemical structure of MKK-1775 is shown below.
Figure imgf000008_0002
Preferred PARP inhibitors are selected from the group consisting of: olaparib, rucaparib, niraparib, talazoparib, veliparib, 3-aminobenzamide, and iniparib. In a particularly preferred embodiment, the PARP inhibitor is olaparib. The chemical structures of each of the preferred PARP inhibitors are shown below:
Figure imgf000008_0003
Olaparib
Figure imgf000009_0002
aazoparb
Veliparib
Figure imgf000009_0001
3-aminobenzamide
Figure imgf000010_0001
Iniparib
In a further preferred embodiment of the invention, the DDRi is used in combination with a DNA damaging agent and/or a DNA damaging procedure. A typically, non-limiting, example of a DNA damaging procedure is radiotherapy.
In a particularly preferred embodiment, the DDRi is used in combination with a DNA damaging agent.
Preferred DNA damaging agents are selected from the group consisting of nucleoside analogues and alkylating agents. A particularly preferred nucleoside analogue is fludarabine. Particularly preferred alkylating agents are selected from the group consisting of chlorambucil, bendamustine, mafosfamide, cyclophosphamide and 4-hydroxy-cyclophosphamide (i.e. the active metabolite of both mafosfamide and, following hepatic activation, cyclophosphamide).
The chemical structures of each of these preferred DNA damaging agents are shown below:
Figure imgf000010_0002
Fludarabine
Figure imgf000011_0001
cyclophosphamide
Figure imgf000012_0001
4-hydroxy-cyclophospham ide
High resolution telomere length analysis is typically measured using a single telomere length analysis (STELA) method, which allows complete resolution of telomere lengths at specific chromosome ends9 11. However, any other method that can measure the full range of telomere length from one telomere repeat to several kb of telomere length may be utilised. In some embodiments, telomere length is detected for a single chromosome. Alternatively, a mean telomere length may be detected for a plurality of different chromosomes and, in such embodiments, an average mean telomere length is calculated. In preferred embodiments, mean telomere length is determined for the XpYp chromosome, either alone or in combination with one or more further chromosomes. In preferred embodiments, the mean telomere length is determined using at least one, any two, any three, any four, any five, any six, any seven, any eight, any nine or all ten of the following chromosomes: XpYp, 17p, 2p, 16p, 18q, 7q, 1 1 q, 9p, 5p and 12q. More preferably, the mean telomere length is determined using at least any one, any two, any three, any four, five or all six of the following chromosomes: XpYp, 17p, 2p, 16p, 18q and 7q chromosomes.
Whilst in principle telomere length can be assessed using any type of cell, the mean telomere length will typically be determined from at least a sample of the cancerous cell type. Therefore, in preferred embodiments, the mean telomere length is determined using at least a sample of B-cells isolated from the patient to be treated.
According to a second aspect of the invention, there is provided a method for treating cancer in a patient, the method comprising: a. using high-resolution telomere length analysis to determine the mean telomere length (in kb) of at one chromosome selected from XpYp, 17p, 2p, 16p, 18q and 7q in at least a sample of cancer cells from said patient; and
b. where said mean telomere length is less than or equal to a threshold value selected according to the chromosome that was tested from the group consisting of:
i. 3.81 kb, preferably 2.26 kb, for chromosome XpYp;
ii. 4.81 kb, preferably 2.57 kb, for chromosome 17p;
iii. 5.01 kb, preferably 3.01 kb, for chromosome 2p;
iv. 4.49 kb, preferably 2.94 kb, for chromosome 16p;
v. 4.47 kb, preferably 2.66 kb, for chromosome 18q;
vi. 4.27 kb, preferably 3.05 kb, for 7q; or
vii. a mean telomere length threshold determined using any two or more of the above threshold values identified in i) to vi) depending upon the combination of different chromosomes that are tested,
c. administering a DDRi to said patient.
As is the case for the first aspect of the invention, the cancer may be any type of cancer but according to a preferred embodiment, the form of cancer is a 13- cell malignant condition. Similarly, in a particularly preferred embodiment, the B-cell malignant condition is CLL, the cancerous cells of which may or may not possess large scale chromosomal abnormalities / deletions, such abnormalities / deletions usually being found in, but not limited to, chromosomes 1 1 q and 17p.
In a preferred embodiment of the second aspect, the DDRi is selected from the group consisting of inhibitors of one or more proteins selected from the group consisting of an ATM, ATR, Chk1 , CHk2, DNA-PKcs, POLQ, WEE1A, WEE1 B, and PARP. More preferably the DDRi is selected from the group consisting of inhibitors of one or more of Chk1 , WEE1A and PARP. In a particularly preferred embodiment, the DDRi is a PARP inhibitor. A preferred Chk1 inhibitor is GDC-0575. A preferred WEE1A inhibitor is MKK- 1775. Preferred PARP inhibitors are selected from the group consisting olaparib, rucaparib, niraparib, talazoparib, veliparib, 3-aminobenzamide, and iniparib. In a particularly preferred embodiment, the PARP inhibitor is olaparib.
In a further preferred embodiment of the invention, the DDRi is administered in combination with a DNA damaging agent and/or before, during or after a DNA damaging procedure. As noted above, a typical, non-limiting, example of a DNA damaging procedure is radiotherapy. In particularly preferred embodiments, the DDRi is administered in combination with a DNA damaging agent.
As noted above, preferred DNA damaging agents are selected from the group consisting of nucleoside analogues and alkylating agents. A particularly preferred nucleoside analogue is fludarabine. Particularly preferred alkylating agents are selected from the group consisting of chlorambucil, bendamustine, mafosfamide, cyclophosphamide and 4-hydroxy-cyclophosphamide.
Again, high resolution telomere length analysis is typically measured using a single telomere length analysis (STELA) method. However, any other method that can measure the full range of telomere length from one telomere repeat to several kb of telomere length may be utilised in the second aspect of the invention. In some embodiments, telomere length is detected for a single chromosome. Alternatively, a mean telomere length may be detected for a plurality of different chromosomes and, in such embodiments, an average mean telomere length is calculated. In a preferred embodiment, mean telomere length is determined for the XpYp chromosome, either alone or in combination with one or more of said further chromosomes. In preferred embodiments, the mean telomere length is determined using at least one, two, three, four, five, six, seven, eight, nine or all ten of the following chromosomes: XpYp, 17p, 2p, 16p, 18q, 7q, 1 1 q, 9p, 5p and 12q. In more preferred embodiments, the mean telomere length is determined using at least one, two, three, four, five or all six of the following chromosomes: XpYp, 17p, 2p, 16p, 18q and 17q chromosomes. Mean telomere length is typically determined from at least a sample of cancer cell type. Therefore, in preferred embodiments, the mean telomere length is determined using at least a sample of B-cells isolated from the patient to be treated.
In the claims which follow and in the preceding description of the invention, except where the context requires otherwise due to express language or necessary implication, the word“comprises”, or variations such as“comprises” or“comprising” is used in an inclusive sense i.e. to specify the presence of the stated features but not to preclude the presence or addition of further features in various embodiments of the invention.
All references, including any patent or patent application, cited in this specification are hereby incorporated by reference. No admission is made that any reference constitutes prior art. Further, no admission is made that any of the prior art constitutes part of the common general knowledge in the art.
Preferred features of each aspect of the invention may be as described in connection with any of the other aspects.
Other features of the present invention will become apparent from the following examples. Generally speaking, the invention extends to any novel one, or any novel combination, of the features disclosed in this specification (including the accompanying claims and drawings). Thus, features, integers, characteristics, compounds or chemical moieties described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein, unless incompatible therewith.
Moreover, unless stated otherwise, any feature disclosed herein may be replaced by an alternative feature serving the same or a similar purpose.
The invention will now be described by way of example only with reference to the following tables and figures:
Table 1. Longest mean telomere length at which telomere end-end fusion events can be detected for a range of chromosomes, including the mean thereof and (derived therefrom) the prognostic mean telomere length for each one of said chromosomes, including the mean thereof. Data adapted from WO 2013/024264.
Table 2. LD50 values for tested individual DNA Damaging agents and DDRis in monoculture conditions.
Figure 1. Comparative cytotoxicity of (A) fludarabine (B) chlorambucil (C) bendamustine (D) mafosfamide (E) rituximab and (F) olaparib in monoculture and co-culture for 48h. The figures show composite dose-response curves for 10 individual patients; all drugs were tested in the same patient samples (n = 10).
Figure 2. Comparison of the cytotoxic effect in primary CLL cell monocultures treated with (A) fludarabine (B) chlorambucil (C) bendamustine (D) mafosfamide (E) olaparib in patient samples grouped according to telomere length.
Figure 3. Comparison of the cytotoxic effect in primary CLL cells co-cultured on CD40L-expressing mouse fibroblasts and treated with (A) fludarabine (B) chlorambucil (C) bendamustine (D) mafosfamide (E) olaparib in patient samples grouped according to telomere length.
Figure 4. Comparison of the synergistic cytotoxic effect in primary CLL cell monocultures treated with a combination of olaparib and (A) fludarabine, (B) chlorambucil, (C) bendamustine, and (D) mafosfamide. The short telomere subset showed significantly enhanced synergy with all four drug combinations in comparison with that observed for the long telomere subset.
Figure 5. Comparison of the synergistic cytotoxic effect in primary CLL cells co-cultured on CD40L-expressing mouse fibroblasts that were treated with a combination of olaparib and (A) fludarabine, (B) chlorambucil, (C) bendamustine, and (D) mafosfamide. The short telomere subset showed strong synergy with all four drug combinations whereas the long telomere subset demonstrated additive effects or weak synergy. Figure 6. Comparison of the synergistic cytotoxic effect in primary CLL cell monocultures treated with a combination of MK-1775 and (A) bendamustine or (B) mafosfamide. The short telomere subset showed significantly enhanced synergy with both drug combinations in comparison with that observed for the long telomere subset.
Figure 7. Comparison of the synergistic cytotoxic effect in primary CLL cell monocultures treated with a combination of GDC-0575 and (A) bendamustine or (B) mafosfamide. The short telomere subset showed significantly enhanced synergy with both drug combinations in comparison with that observed for the long telomere subset.
METHODS
CLL Patients
Peripheral blood samples from CLL consenting patients, in accordance with the Declaration of Helsinki and as approved by the South East Wales local research ethics committee (LREC# 02/4806). CLL was defined by clinical criteria as well as cellular morphology, and also the co-expression of CD19 and CD5 in lymphocytes simultaneously displaying restriction of light-chain rearrangement. All of the samples were collected at, or close to, the time of diagnosis and staging was based on the Binet classification system16.
Isolation of peripheral blood mononuclear cells from CLL patients
Peripheral blood mononuclear cells (PBMCs) were isolated from EDTA venous blood of CLL patients by density centrifugation using Ficoll-Hypaque (Invitrogen).
Purification of CLL B-cells
CD19+ B-cells were positively selected by using CD19+ dynabeads from Life Technologies. 10pL of the desired bead was used for the isolation of 500,000 target cells form isolated PBMCs. The cell preparation containing the cells of interest and the washed dynabeads was incubated for 20 minutes at 4°C. Cells were isolated by placing the microtube back into the magnetic particle concentrator for 2 minutes. After this time, the supernatant was aspirated and discarded and the microtube was removed from the magnetic particle concentrator.
Telomere length analysis using STELA
DNA was extracted using the Maxwell® 16 Blood DNA Purification Kit on the Maxwell® 16 Instrument (Promega, Madison, Wl, USA). For telomere length analysis at the XpYp telomere we used an adaptation of the chromosome- specific single telomere length analysis (STELA) assay, as previously described9 11, to allow for high-throughput analysis (HT-STELA). In particular, the STELA protocol was adapted to use telomere-adjacent primers specific for the XpYp telomere (XpYpC: 5' -C AG G G AC C G G GAC AAAT AG AC-3' ) , in triplicate 30pl PCR reactions each containing 30 ng of genomic DNA. Thermal cycling conditions were: 23 cycles of 94°C for 20 s, 65°C for 30 s and 68°C for 5 mins. Amplified fragments were resolved using capillary gel electrophoresis and mean telomere length determined using PROSize software (AATI, Ankeny, Iowa, USA).
CLL PBMC Cultures
Primary CLL cells have the propensity to die very quickly once removed from the patient. However, it has previously been shown that such‘spontaneous apoptosis’ can be inhibited by the addition of interleukin 4 (IL-4) into the culture media17. Therefore, all culture data generated in this report was obtained using Roswell Park Memorial Institute (RPMI) 1640 (Sigma) culture media supplemented with 5ng/mL interleukin 4 (IL-4).
The cytotoxicity of all the agents were first tested in the following monoculture conditions: 1x106 CLL cells were resuspended in 1 mL of cell culture medium - RPMI (1640 Sigma), which was supplemented with: 2mM L-Glutamine (Life Technologies); 100units/ml Penicillin and 100g/mL streptomycin (Life Technologies); and 10% foetal Calf Serum (FCS) (Life Technologies) supplemented with 5ng/pL IL-4 (Biosource).
In addition to monoculture experiments, we also tested the cytotoxicity of all the agents in CD40L co-culture: CD40L-expressing mouse fibroblast cells were suspended in 5m L of fibroblast media and irradiated (8000RADs). 1x105 irradiated cells were then seeded into a 24-well plate. The plate was incubated overnight at 37°C with 5% CO2 to allow the fibroblast cells to adhere to the well surface. Subsequently, duplicate aliquots of 1x106 CLL patient’s PBMCs were taken and re-suspended in cell culture medium (3ml) - RPMI as above, which was supplemented with: 2mM L-Glutamine (Life Technologies); 100units/ml Penicillin and 100g/mL streptomycin (Life Technologies); and 10% FCS (Life Technologies) supplemented with 5ng/pL IL-4 (Biosource). These samples were left to incubate at 37°C in a 5% CO2 moist chamber for 1 hour. The 1x106 CLL patient’s PBMCs were then added to the surface of the irradiated cells and incubated at 37°C in a 5% CO2 humidified chamber.
On day 3 or 4, 1 mL of fresh culture medium was added to each sample well. Every 7 days the cells were transferred to new CD40L feeder wells. We have previously shown that co-culturing CLL cells in this way causes CLL cell activation and proliferation17.
RESULTS
The in vitro cytotoxicity of a range of DNA damaging agents was assessed in monoculture (i.e. CLL cells alone) and co-culture (i.e. CLL cells on CD40L- expressing mouse fibroblasts) to determine whether sensitivity to these agents was related to the telomere length profiles of the tumour cells. Subsequently, these agents were combined with various DDRi compounds, in particular olaparib (a PARP inhibitor), GDC-0575 (a Chk1 inhibitor) and MKK-1775 (a WEE1A inhibitor), in the same in vitro models. Analysis of the efficacy of chemotherapy agents, rituximab and the PARP inhibitor, olaparib, to kill primary CLL cells in monoculture and CD40L- expressing co-culture
We have analysed the comparative cytotoxicity of fludarabine, chlorambucil, bendamustine, mafosfamide, rituximab and olaparib as single agents in both a monoculture system and a co-culture system (Figure 1 ).
Our results demonstrate that the purine nucleoside analogue (fludarabine) and the three alkylating agents (chlorambucil, bendamustine and mafosfamide) all show reduced cytotoxicity under co-culture conditions, and rituximab showed little cytotoxicity under either monoculture or co-culture conditions.
In contrast, olaparib was shown to enhance the cytotoxic effect when applied to CLL cells in co-culture. Although not bound to any particular theory (at least because the mechanism for this increased potency was not explored in this study), one may speculate that this increased cellular activation and tumour cell division induced by the co-culture conditions may make cells more prone to DNA damage and hence more reliant on DNA repair mechanisms. Therefore, DDRis such as PARP inhibitors may, even as monotherapies, be more effective in proliferating cells within a tumour.
The impact of telomere length on the ability of chemotherapy agents, rituximab and olaparib to kill primary CLL cells in monoculture and coculture
To assess if there was any relationship between telomere length and the cytotoxic response to drugs, the telomere length distribution in our CLL patient samples was analysed using single telomere length analysis (STELA) at the XpYp telomere, and the patients were divided into two groups based on telomere length (i.e. telomere length below 3.81 kb or a telomere length at/above 3.81 kb, 3.81 kb representing the upper threshold at which telomere end-end fusion events can be detected) before the response to drugs in our in vitro models was compared between said groups (Figure 2 and Figure 3).
These results demonstrated that the short telomere group was significantly resistant to killing with the purine nucleoside analogue (fludarabine) and the three alkylating agents (chlorambucil, bendamustine and mafosfamide) in both monoculture (Figure 2A-2D) and co-culture (Figure 3A-3D). In contrast, the same cells showed increased sensitivity to olaparib under both sets of in vitro conditions (Figure 2E and Figure 3E respectively).
It was also noted that CLL cells derived from patients harbouring an 1 1 q deletion or a 17p deletion showed no significant difference in response to the drugs when the samples were grouped according to telomere length. This implies that CLL cells with short telomeres are inherently more resistant to the cytotoxic effect of DNA damaging agents, and more sensitive to the cytotoxic effects of DDRis such as PARP inhibitors, regardless of their cytogenetic background.
Analysis of the cytotoxic efficacy of chemotherapy agents and rituximab, when used in combination with olaparib, against primary CLL cells
To assess if there was any antagonistic, additive or synergistic effect upon the cytotoxic efficacy of DNA damaging agents, the experimentally determined LD50 values of each of the individual agents (Table 2) from the monoculture studies detailed above were used to determine the molar ratios for the combination studies. Specifically, potential synergistic enhancement of cytotoxic efficacy was evaluated in both monoculture (Figure 4) and co-culture (Figure 5) experiments using the following DNA Damaging AgentDDRi molar ratios:
• 1 : 1 for chlorambucil:olaparib combinations;
• 1 :2 for bendamustine:olaparib combinations;
• 1 :40 for fludarabine:olaparib combinations; and
• 1 : 100 for mafosfamide combinations. These results not only demonstrate that the PARP inhibitor olaparib was synergistic with fludarabine, chlorambucil, bendamustine and mafosfamide (particularly in DNA Damaging Agent: DDRi molar ratios ranging from 1000: 1 to 1 :1000, preferably 500: 1 to 1 : 1000, more preferably 10: 1 to 1 :1000, and still more preferably from 5:1 to 1 :500), but also that a significantly stronger synergy was observed for the short telomere patient group (Figures 4 and 5).
Analysis of the cytotoxic efficacy of chemotherapy agents when used in combination with WEE1A inhibitors or Chk1 inhibitors, against primary CLL cells
To assess if the synergistic effect upon the cytotoxic efficacy of DNA damaging agents for combinations of DNA damaging agents and DDRis was specific to combinations comprising PARP inhibitor DDRis, cytotoxicity efficacy was also evaluated in monoculture experiments using combinations comprising the WEE1A inhibitor MK-1775 (Figure 6) or the Chk1 inhibitor GDC-0575 (Figure 7) using the following DNA Damaging Agent: DDRi molar ratios:
• 500: 1 for bendamustine:MKK-1775 and bendamustine:GDC-0575 combinations; and
• 50: 1 for mafosfamide:MKK-1775 and bendamustine combinations.
Consistent with the results obtained using combinations comprising the PARP inhibitor olaparib, these results not only demonstrate that the WEE1A inhibitor MKK-1775 and Chk1 inhibitor GDC-0575 were each synergistic with the DNA damaging agents bendamustine and mafosfamide, but also that a significantly stronger synergy was observed for the short telomere patient group. In particular, these synergistic effects were shown for combinations comprising DNA Damaging Agent: DDRi molar ratios ranging from 1000: 1 to 1 :1000, more preferably from 750: 1 to 25: 1 and still more preferably from 500: 1 to 50: 1 . Summary
The main findings of this study can be summarised as follows:
The work presented here provides a rationale for using DDRis such as PARP inhibitors, WEE1A inhibitors or Chk1 inhibitors, alone or in combination with DNA damaging agents/procedures, for the treatment of cancer.
The enhanced synergistic efficacy observed in the short telomere group suggests that tumour cells with short telomeres are inherently more sensitive to inhibition of DNA damage response pathways.
It is also noted that short telomeres are associated with genomic complexity in a number of human cancers. Therefore, one can speculate that patients with these adverse clinical characteristics may preferentially benefit from the combination of DNA damage response pathway inhibitors and DNA damaging agents.
Consequently, tumour cell telomere length potentially could be used to risk- stratify patients (routinely or in clinical trials) and assign them to drug treatment regimens specifically designed to exploit their telomere length-associated weakness.
References
1. Gertler R, Rosenberg R, Strieker D, et al. Telomere length and human telomerase reverse transcriptase expression as markers for progression and prognosis of colorectal carcinoma. J Clin Oncol 2004;22:1807-14.
2. Meeker AK, Argani P. Telomere shortening occurs early during breast tumorigenesis: a cause of chromosome destabilization underlying malignant transformation? J Mammary Gland Biol Neoplasia 2004;9:285-96.
3. Damle RN, Batliwalla FM, Ghiotto F, et al. Telomere length and telomerase activity delineate distinctive replicative features of the B-CLL subgroups defined by immunoglobulin V gene mutations. Blood 2004;103:375-82.
4. Grabowski P, Hultdin M, Karlsson K, et al. Telomere length as a prognostic parameter in chronic lymphocytic leukemia with special reference to VH gene mutation status. Blood 2005;105:4807-12.
5. Rossi D, Lobetti Bodoni C, Genuardi E, et al. Telomere length is an independent predictor of survival, treatment requirement and Richter's syndrome transformation in chronic lymphocytic leukemia. Leukemia 2009;23:1062-72.
6. Sellmann L, de Beer D, Bartels M, et al. Telomeres and prognosis in patients with chronic lymphocytic leukaemia. Int J Hematol 2011 ;93:74-82.
7. Roos G, Krober A, Grabowski P, et al. Short telomeres are associated with genetic complexity, high risk genomic aberrations, and short survival in chronic lymphocytic leukemia. Blood 2008; 1 11 :2246-52.
8. Ricca I, Rocci A, Drandi D, et al. Telomere length identifies two different prognostic subgroups among VH-unmutated B-cell chronic lymphocytic leukemia patients. Leukemia 2007; 21 :697-705.
9. Baird DM, Rowson J, Wynford-Thomas D, Kipling D. Extensive allelic variation and ultrashort telomeres in senescent human cells. Nat Genet. 2003; 33:203-7.
10. Britt-Compton B, Rowson J, Locke M, Mackenzie I, Kipling D, Baird DM. Structural stability and chromosome-specific telomere length is governed by cis-acting determinants in humans. Hum Mol Genet. 2006; 15:725-33.
11. Capper R, Britt-Compton B, Tankimanova M, et al. The nature of telomere fusion and a definition of the critical telomere length in human cells. Genes Dev. 2007; 21 :2495- 2508. Letsolo BT, Rowson J, Baird DM. Fusion of short telomeres in human cells is characterised by extensive deletion and microhomology and can result in complex rearrangements. Nucleic Acids Res. 2010; 38:1841 -52.
Lin TT, Norris K, Heppel NH, Pratt G, Allan JM, Allsup DJ, Bailey J, Cawkwell L, Hills R, Grimstead JW, Jones RE, Britt-Compton B, Fegan C, Baird DM, Pepper C. Telomere dysfunction accurately predicts clinical outcome in chronic lymphocytic leukaemia, even in patients with early stage disease. Br J Haematol. 2014 Oct;167(2):214-223.
Strefford JC, Kadalayil L, Forster J, Rose-Zerilli MJ, Parker A, Lin TT, Heppel N, Norris K, Gardiner A, Davies Z, Gonzalez de Castro D, Else M, Steele AJ, Parker H, Stankovic T, Pepper C, Fegan C, Baird D, Collins A, Catovsky D, Oscier DG. Telomere length predicts progression and overall survival in chronic lymphocytic leukemia: data from the UK LRF CLL4 trial. Leukemia. 2015 Dec; 29(12):241 1 -2414.
Norris K, Hillmen P, Rawstron A, Hills R, Baird DM, Fegan CD, Pepper C. Telomere length predicts for outcome to FCR chemotherapy in CLL. In submission.
Binet JL, Auquier A, Dighiero G, et al. A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer 1981 ; 48: 198-206.
Hamilton E, Pearce L, Morgan L, Robinson S, Ware V, Brennan P, Thomas NS, Yallop D, Devereux S, Fegan C, Buggins AG, Pepper C. Mimicking the tumour microenvironment: three different co-culture systems induce a similar phenotype but distinct proliferative signals in primary chronic lymphocytic leukaemia cells. Br J Haematol. 2012 Sep;158(5):589-599.
Table 1. Upper limit and mean telomere length at which telomere end-end fusion events can be detected for five different chromosome ends
Figure imgf000026_0001
value calculated based on XpYp, 17p, 2p, 16p and 18q determinations only
Table 2. LD50 values for tested individual DNA Damaging agents and DDRis in monoculture conditions
Figure imgf000026_0002

Claims

Claims
1 . A DNA damage response inhibitor (DDRi) for use in treating cancer in a patient whose cancer cells have a mean telomere length, determined using high-resolution telomere length analysis, that is less than or equal to a threshold value selected according to the chromosome to be tested from the group consisting of: i) 3.81 kb for XpYp;
ii) 4.81 kb for 17p;
iii) 5.01 kb for 2p;
iv) 4.49 kb for 16p;
v) 4.47 kb for 18q;
vi) 4.27 kb for 7q; or
vii) A mean telomere length threshold determined using any two or more of the above threshold values identified in i) to vi) depending upon the combination of different chromosomes that are tested.
2. The DDRi for use according to claim 1 , wherein the mean telomere length of said cells is determined as the average of the mean telomere lengths determined for each of XpYp, 17p, 2p, 16p and 18q, and wherein said threshold value is 4.52 kb.
3. The DDRi for use according to claim 1 or claim 2, wherein said cancer is a B-cell malignant condition.
4. The DDRi for use according to claim 3, wherein said B-cell malignant condition is chronic lymphocytic leukaemia (CLL).
5. The DDRi for use according to claim 4, wherein said cancer cells are free of chromosomal abnormalities (cytogenetically normal).
6. The DDRi for use according to 4, wherein said cancer cells possess one or more chromosomal abnormalities.
7. The DDRi for use according to claim 6, wherein said chromosomal abnormalities are selected from deletions in one or more chromosomes selected from the group consisting of 1 1 q and 17p.
8. The DDRi for use according to any one of claims 1 -7, wherein said DDRi is an inhibitor of one or more proteins selected from the group consisting of: an ATM serine/threonine kinase (ATM), ATR serine/threonine- protein kinase (ATR), Checkpoint kinase 1 (Chk1 ), Checkpoint kinase 2 (CHk2), DNA-dependent protein kinase catalytic subunit (DNA-PKcs), DNA polymerase theta (POLQ), Wee1 -like protein kinase (WEE1A), Wee1 -like protein kinase 2 (WEE1 B), and poly ADP ribose polymerase (PARP).
9. The DDRi for use according to claim 8, wherein said DDRi is an inhibitor of one or more proteins selected from the group consisting of WEE1 A, Chk1 and PARP.
10. The DDRi for use according to claim 9, wherein:
a. said DDRi is a WEE1A inhibitor, optionally MKK-1775; or b. said DDRi is a Chk1 inhibitor, optionally GDC-0575.
1 1 . The DDRi for use according to claim 9, wherein said DDRi is a PARP inhibitor optionally selected from the group consisting of: olaparib, rucaparib, niraparib, talazoparib, veliparib, 3-aminobenzamide, and iniparib.
12. The DDRi for use according to claim 1 1 , wherein said PARP inhibitor is olaparib.
13. The DDRi for use according to any one of claims 1 -12, wherein said DDRi is used in combination with a DNA damaging agent and/or a DNA damaging procedure.
14. The DDRi for use according to claim 13, wherein said DNA damaging agent is selected from the group consisting of nucleoside analogues and alkylating agents.
15. The DDRi for use according to claim 14, wherein said nucleoside analogue is fludarabine, and/or wherein said alkylating agent is selected from the group consisting of: chlorambucil, bendamustine, mafosfamide, cyclophosphamide and 4-hydroxy-cyclophosphamide.
16. The DDRi for use according to any one of claims 13-15, wherein said DNA damaging procedure is radiotherapy.
17. The DDRi for use according to any one of claims 1 -16, wherein said mean telomere length is determined in at least a sample of B-cells isolated from said patient.
18. The DDRi for use according to any one of claims 1 -17, wherein said mean telomere length is determined for a single chromosome.
19. The DDRi for use according to any one of claims 1 -17, wherein said mean telomere length is determined for a plurality of different chromosomes.
20. The DDRi for use according to any one of claims 1 -19, wherein said mean telomere length is determined for chromosome XpYp.
21.A method for treating cancer in a patient, the method comprising: a. using high-resolution telomere length analysis to determine the mean telomere length (in kb) of at one chromosome selected from XpYp, 17p, 2p, 16p, 18q and 7q in at least a sample of cancer cells from said patient; and
b. where said mean telomere length is less than or equal to a threshold value selected according to the chromosome that was tested from the group consisting of:
i. 3.81 kb for chromosome XpYp;
ii. 4.81 kb for chromosome 17p;
iii. 5.01 kb for chromosome 2p;
iv. 4.49 kb for chromosome 16p;
v. 4.47 kb for chromosome 18q;
vi. 4.27 kb for 7q; or
vii. a mean telomere length threshold determined using any two or more of the above threshold values identified in i) to vi) depending upon the combination of different chromosomes that are tested,
c. administering a DDRi to said patient.
22. The method according to claim 21 , wherein the mean telomere length of said cells is determined as the average of the mean telomere lengths determined for each of XpYp, 17p, 2p, 16p and 18q, and wherein said threshold value is 4.52 kb.
23. The method according to claim 21 or claim 22, wherein said cancer is a B-cell malignant condition.
24. The method according to claim 23, wherein said B-cell malignant condition is chronic lymphocytic leukemia (CLL).
25. The method according to claim 24, wherein said cancerous cells are free of chromosomal abnormalities (cytogenetically normal).
26. The method according to 24, wherein said cancerous cells possesses one or more chromosomal abnormalities.
27. The method according to claim 26, wherein said chromosomal abnormalities are selected from deletions in one or more chromosomes selected from the group consisting of 11 q and 17p.
28. The method according to any one of claims 21 -27, wherein said DDRi is an inhibitor of one or more proteins selected from the group consisting of: an ATM serine/threonine kinase (ATM), ATR serine/threonine- protein kinase (ATR), Checkpoint kinase 1 (Chk1 ), Checkpoint kinase 2 (CHk2), DNA-dependent protein kinase catalytic subunit (DNA-PKcs), DNA polymerase theta (POLQ), Wee1 -like protein kinase (WEE1A), Wee1 -like protein kinase 2 (WEE1 B), and poly ADP ribose polymerase (PARP).
29. The method according to claim 28, wherein said DDRi is an inhibitor of one or more proteins selected from the group consisting of WEE1A, Chk1 and PARP.
30. The method according to claim 29, wherein:
a. Said DDRi is a WEE1 inhibitor, optionally MKK-1775; or b. said DDRi is a Chk1 inhibitor, optionally GDC-0575.
31. The method according to claim 29, wherein said DDRi is a PARP inhibitor optionally selected from the group consisting of: olaparib, rucaparib, niraparib, talazoparib, veliparib, 3-aminobenzamide, and iniparib.
32. The method according to claim 31 , wherein said PARP inhibitor is olaparib.
33. The method according to any one of claims 21 -33, wherein said DDRi is administered in combination with a DNA damaging agent and/or a DNA damaging procedure.
34. The method according to claim 33, wherein said DNA damaging agent is selected from the group consisting of nucleoside analogues and alkylating agents.
35. The method according to claim 34, wherein said nucleoside analogue is fludarabine, and/or wherein said alkylating agent is selected from the group consisting of: chlorambucil, bendamustine, mafosfamide, cyclophosphamide and 4-hydroxy-cyclophosphamide.
36. The method according to any one of claims 33-35, wherein said DNA damaging procedure is radiotherapy.
37. The method according to any one of claims 21 -36, wherein said mean telomere length is determined in at least a sample of B-cells isolated from said patient.
38. The method according to any one of claims 21 -37, wherein said mean telomere length is determined for a single chromosome.
39. The method according to any one of claims 21 -37, wherein said mean telomere length is detected for a plurality of different chromosomes.
40. The method according to any one of claims 21 -39, wherein said mean telomere length is detected for chromosome XpYp.
PCT/GB2019/053261 2018-11-19 2019-11-18 Compounds for telomere length-related treatment of cancer WO2020104777A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1818792.2 2018-11-19
GBGB1818792.2A GB201818792D0 (en) 2018-11-19 2018-11-19 Compounds for telomere length-related treatment of cancer

Publications (1)

Publication Number Publication Date
WO2020104777A1 true WO2020104777A1 (en) 2020-05-28

Family

ID=64740185

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2019/053261 WO2020104777A1 (en) 2018-11-19 2019-11-18 Compounds for telomere length-related treatment of cancer

Country Status (2)

Country Link
GB (1) GB201818792D0 (en)
WO (1) WO2020104777A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006125166A2 (en) * 2005-05-18 2006-11-23 Alt Solutions, Inc. Pharmacological modulation of telomere length in cancer cells for prevention and treatment of cancer
KR20120136686A (en) * 2011-06-09 2012-12-20 주식회사 디앤피바이오텍 Method for predicting prognosis in patients with non-small cell lung cancer using telomere shortening
WO2013024264A1 (en) 2011-08-15 2013-02-21 University College Cardiff Consultants Limited Prognostic methodology
WO2014085632A1 (en) * 2012-11-30 2014-06-05 Geron Corporation Diagnostic markers for treating cell proliferative disorders with telomerase inhibitors
US20140287409A1 (en) * 2013-03-15 2014-09-25 Georgetown University Detection And Assessment Of Cancer Risk Using Telomere Health
WO2016059398A1 (en) * 2014-10-14 2016-04-21 University College Cardiff Consultants Limited High throughput sequencing
WO2016149618A1 (en) * 2015-03-19 2016-09-22 The Johns Hopkins University Method for regulation of telomere-length

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006125166A2 (en) * 2005-05-18 2006-11-23 Alt Solutions, Inc. Pharmacological modulation of telomere length in cancer cells for prevention and treatment of cancer
KR20120136686A (en) * 2011-06-09 2012-12-20 주식회사 디앤피바이오텍 Method for predicting prognosis in patients with non-small cell lung cancer using telomere shortening
WO2013024264A1 (en) 2011-08-15 2013-02-21 University College Cardiff Consultants Limited Prognostic methodology
WO2014085632A1 (en) * 2012-11-30 2014-06-05 Geron Corporation Diagnostic markers for treating cell proliferative disorders with telomerase inhibitors
US20140287409A1 (en) * 2013-03-15 2014-09-25 Georgetown University Detection And Assessment Of Cancer Risk Using Telomere Health
WO2016059398A1 (en) * 2014-10-14 2016-04-21 University College Cardiff Consultants Limited High throughput sequencing
WO2016149618A1 (en) * 2015-03-19 2016-09-22 The Johns Hopkins University Method for regulation of telomere-length

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
BAIRD DMROWSON JWYNFORD-THOMAS DKIPLING D: "Extensive allelic variation and ultrashort telomeres in senescent human cells", NAT GENET., vol. 33, 2003, pages 203 - 7, XP002246105, DOI: 10.1038/ng1084
BINET JLAUQUIER ADIGHIERO G ET AL.: "A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis", CANCER, vol. 48, 1981, pages 198 - 206
BRITT-COMPTON BROWSON JLOCKE MMACKENZIE IKIPLING DBAIRD DM: "Structural stability and chromosome-specific telomere length is governed by cis-acting determinants in humans", HUM MOL GENET., vol. 15, 2006, pages 725 - 33, XP055614328, DOI: 10.1093/hmg/ddi486
CAPPER RBRITT-COMPTON BTANKIMANOVA M ET AL.: "The nature of telomere fusion and a definition of the critical telomere length in human cells", GENES DEV., vol. 21, 2007, pages 2495 - 2508
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1196541-47-5
D ROSSI ET AL: "Telomere length is an independent predictor of survival, treatment requirement and Richter's syndrome transformation in chronic lymphocytic leukemia", LEUKEMIA., vol. 23, no. 6, 2 April 2009 (2009-04-02), US, pages 1062 - 1072, XP055592946, ISSN: 0887-6924, DOI: 10.1038/leu.2008.399 *
DAMLE RNBATLIWALLA FMGHIOTTO F ET AL.: "Telomere length and telomerase activity delineate distinctive replicative features of the B-CLL subgroups defined by immunoglobulin V gene mutations", BLOOD, vol. 103, 2004, pages 375 - 82
DONA FRANCESCA ET AL: "Poly(ADP-ribosylation) and Neoplastic Transformation: Effect of PARP Inhibitors", vol. 14, no. 5, 31 May 2013 (2013-05-31), pages 524 - 536, XP009513580, ISSN: 1389-2010, Retrieved from the Internet <URL:1077952576> DOI: 10.2174/138920101405131111104642 *
GERTLER RROSENBERG RSTRICKER D ET AL.: "Telomere length and human telomerase reverse transcriptase expression as markers for progression and prognosis of colorectal carcinoma", J CLIN ONCOL, vol. 22, 2004, pages 1807 - 14
GRABOWSKI PHULTDIN MKARLSSON K ET AL.: "Telomere length as a prognostic parameter in chronic lymphocytic leukemia with special reference to VH gene mutation status", BLOOD, vol. 105, 2005, pages 4807 - 12
GURUNG RESHAM LAL ET AL: "Targeting DNA-PKcs and telomerase in brain tumour cells", MOLECULAR CANCER, vol. 13, 13 October 2014 (2014-10-13), XP002791725, ISSN: 1476-4598 *
HAMILTON EPEARCE LMORGAN LROBINSON SWARE VBRENNAN PTHOMAS NSYALLOP DDEVEREUX SFEGAN C: "Mimicking the tumour microenvironment: three different co-culture systems induce a similar phenotype but distinct proliferative signals in primary chronic lymphocytic leukaemia cells", BR J HAEMATOL., vol. 158, no. 5, September 2012 (2012-09-01), pages 589 - 599
LETSOLO BTROWSON JBAIRD DM: "Fusion of short telomeres in human cells is characterised by extensive deletion and microhomology and can result in complex rearrangements", NUCLEIC ACIDS RES., vol. 38, 2010, pages 1841 - 52
LIN T T ET AL: "Telomere dysfunction accurately predicts clinical outcome in chronic lymphocytic leukaemia, even in patients with early stage disease", BRITISH JOURNAL OF HAEMATOLOGY, WILEY-BLACKWELL PUBLISHING LTD, GB, vol. 167, no. 2, 1 October 2014 (2014-10-01), pages 214 - 223, XP002751658, ISSN: 0007-1048, [retrieved on 20140703], DOI: 10.1111/BJH.13023 *
LIN TTNORRIS KHEPPEL NHPRATT GALLAN JMALLSUP DJBAILEY JCAWKWELL LHILLS RGRIMSTEAD JW: "Telomere dysfunction accurately predicts clinical outcome in chronic lymphocytic leukaemia, even in patients with early stage disease", BR J HAEMATOL., vol. 167, no. 2, October 2014 (2014-10-01), pages 214 - 223, XP002751658, DOI: 10.1111/bjh.13023
MEEKER AKARGANI P: "Telomere shortening occurs early during breast tumorigenesis: a cause of chromosome destabilization underlying malignant transformation?", J MAMMARY GLAND BIOL NEOPLASIA, vol. 9, 2004, pages 285 - 96
MIRJOLET CELINE ET AL: "The role of telomeres in predicting individual radiosensitivity of patients with cancer in the era of personalized radiotherapy", CANCER TREATMENT REVIEWS, vol. 41, no. 4, April 2015 (2015-04-01), pages 354 - 360, XP002791726 *
NORRIS KHILLMEN PRAWSTRON AHILLS RBAIRD DMFEGAN CDPEPPER C, TELOMERE LENGTH PREDICTS FOR OUTCOME TO FCR CHEMOTHERAPY IN CLL
RICCA IROCCI ADRANDI D ET AL.: "Telomere length identifies two different prognostic subgroups among VH-unmutated B-cell chronic lymphocytic leukemia patients", LEUKEMIA, vol. 21, 2007, pages 697 - 705
ROOS GKROBER AGRABOWSKI P ET AL.: "Short telomeres are associated with genetic complexity, high risk genomic aberrations, and short survival in chronic lymphocytic leukemia", BLOOD, vol. 111, 2008, pages 2246 - 52
ROSSI DLOBETTI BODONI CGENUARDI E ET AL.: "Telomere length is an independent predictor of survival, treatment requirement and Richter's syndrome transformation in chronic lymphocytic leukemia", LEUKEMIA, vol. 23, 2009, pages 1062 - 72, XP055592946, DOI: 10.1038/leu.2008.399
SELLMANN LDE BEER DBARTELS M ET AL.: "Telomeres and prognosis in patients with chronic lymphocytic leukaemia", INT J HEMATOL, vol. 93, 2011, pages 74 - 82
STREFFORD JCKADALAYIL LFORSTER JROSE-ZERILLI MJPARKER ALIN TTHEPPEL NNORRIS KGARDINER ADAVIES Z: "Telomere length predicts progression and overall survival in chronic lymphocytic leukemia: data from the UK LRF CLL4 trial", LEUKEMIA, vol. 29, no. 12, December 2015 (2015-12-01), pages 2411 - 2414
THET THET LIN ET AL: "Telomere dysfunction and fusion during the progression of chronic lymphocytic leukaemia: evidence for a telomere crisis", vol. 116, no. 11, 16 September 2010 (2010-09-16), pages 1899 - 907, XP002687450, ISSN: 0006-4971, Retrieved from the Internet <URL:http://bloodjournal.hematologylibrary.org/content/116/11/1899> [retrieved on 20100610], DOI: 10.1182/BLOOD-2010-02-272104 *

Also Published As

Publication number Publication date
GB201818792D0 (en) 2019-01-02

Similar Documents

Publication Publication Date Title
US20230233563A1 (en) Compounds with anti-tumor activity against cancer cells bearing egfr or her2 exon 20 mutations
Hassler et al. Insights into the pathogenesis of anaplastic large-cell lymphoma through genome-wide DNA methylation profiling
Bolzán et al. DNA and chromosome damage induced by bleomycin in mammalian cells: An update
Wu et al. Genomics and pharmacogenomics of pediatric acute lymphoblastic leukemia
Melo et al. Chronic myeloid leukaemia as a model of disease evolution in human cancer
Hiyama et al. Correlating telomerase activity levels with human neuroblastoma outcomes
JP6995091B2 (en) Methods and kits for screening cancer patients
KR20180134347A (en) Diagnosis and Treatment of Cancer
US11859252B2 (en) Diagnostic and therapeutic methods for cancer
Farooqi et al. Alternative lengthening of telomeres in neuroblastoma cell lines is associated with a lack of MYCN genomic amplification and with p53 pathway aberrations
US11946053B2 (en) Methods for overcoming glucocorticoid resistance and for determining glucocorticoid resistance potential in cancer
JP2022527788A (en) Compounds with antitumor activity against cancer cells with EGFR or HER2 exon 20 insertion
Pellagatti et al. Recent advances in the 5q-syndrome
US20200054660A1 (en) Dna methylation profiling for t-cell immunotherapy
Humphries et al. Pro‐apoptotic TP 53 homolog TA p63 is repressed via epigenetic silencing and B‐cell receptor signalling in chronic lymphocytic leukaemia
US20160194718A1 (en) Compositions and Methods for Identification, Assessment, Prevention, and Treatment of Cancer Using Histone H3K27ME3 Biomarkers and Modulators
Narayan et al. PCDH10 promoter hypermethylation is frequent in most histologic subtypes of mature lymphoid malignancies and occurs early in lymphomagenesis
Ravoet et al. 6q-is an early and persistent chromosomal aberration in CD3-CD4+ T-cell clones associated with the lymphocytic variant of hypereosinophilic syndrome
Remes et al. Telomere length and telomerase activity in malignant lymphomas at diagnosis and relapse
JP2022529016A (en) Tyrosine kinase inhibitor resistant Compounds with antitumor activity against cancer cells with EGFR mutations
Takita Molecular basis and clinical features of neuroblastoma
Alvino et al. A single cycle of treatment with temozolomide, alone or combined with O6-benzylguanine, induces strong chemoresistance in melanoma cell clones in vitro: role of O6-methylguanine-DNA methyltransferase and the mismatch repair system
KR20210145161A (en) Compounds with antitumor activity against cancer cells with HER2 exon 21 insert
CN112088000A (en) Compounds having antitumor activity against cancer cells carrying the HER2 exon 19 mutation
WO2020104777A1 (en) Compounds for telomere length-related treatment of 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: 19806313

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: 19806313

Country of ref document: EP

Kind code of ref document: A1