Search This Blog

Friday, June 16, 2023

Oncogenomics

From Wikipedia, the free encyclopedia
https://en.wikipedia.org/wiki/Oncogenomics

Oncogenomics is a sub-field of genomics that characterizes cancer-associated genes. It focuses on genomic, epigenomic and transcript alterations in cancer.

Cancer is a genetic disease caused by accumulation of DNA mutations and epigenetic alterations leading to unrestrained cell proliferation and neoplasm formation. The goal of oncogenomics is to identify new oncogenes or tumor suppressor genes that may provide new insights into cancer diagnosis, predicting clinical outcome of cancers and new targets for cancer therapies. The success of targeted cancer therapies such as Gleevec, Herceptin and Avastin raised the hope for oncogenomics to elucidate new targets for cancer treatment.

Overall goals of oncogenomics

Besides understanding the underlying genetic mechanisms that initiate or drive cancer progression, oncogenomics targets personalized cancer treatment. Cancer develops due to DNA mutations and epigenetic alterations that accumulate randomly. Identifying and targeting the mutations in an individual patient may lead to increased treatment efficacy.

The completion of the Human Genome Project facilitated the field of oncogenomics and increased the abilities of researchers to find oncogenes. Sequencing technologies and global methylation profiling techniques have been applied to the study of oncogenomics.

History

The genomics era began in the 1990s, with the generation of DNA sequences of many organisms. In the 21st century, the completion of the Human Genome Project enabled the study of functional genomics and examining tumor genomes. Cancer is a main focus.

The epigenomics era largely began more recently, about 2000. One major source of epigenetic change is altered methylation of CpG islands at the promoter region of genes (see DNA methylation in cancer). A number of recently devised methods can assess the DNA methylation status in cancers versus normal tissues. Some methods assess methylation of CpGs located in different classes of loci, including CpG islands, shores, and shelves as well as promoters, gene bodies, and intergenic regions. Cancer is also a major focus of epigenetic studies.

Access to whole cancer genome sequencing is important to cancer (or cancer genome) research because:

  • Mutations are the immediate cause of cancer and define the tumor phenotype.
  • Access to cancerous and normal tissue samples from the same patient and the fact that most cancer mutations represent somatic events, allow the identification of cancer-specific mutations.
  • Cancer mutations are cumulative and sometimes are related to disease stage. Metastasis and drug resistance are distinguishable.

Access to methylation profiling is important to cancer research because:

  • Epi-drivers, along with Mut-drivers, can act as immediate causes of cancers
  • Cancer epimutations are cumulative and sometimes related to disease stage

Whole genome sequencing

The first cancer genome was sequenced in 2008. This study sequenced a typical acute myeloid leukaemia (AML) genome and its normal counterpart genome obtained from the same patient. The comparison revealed ten mutated genes. Two were already thought to contribute to tumor progression: an internal tandem duplication of the FLT3 receptor tyrosine kinase gene, which activates kinase signaling and is associated with a poor prognosis and a four base insertion in exon 12 of the NPM1 gene (NPMc). These mutations are found in 25-30% of AML tumors and are thought to contribute to disease progression rather than to cause it directly.

The remaining 8 were new mutations and all were single base changes: Four were in families that are strongly associated with cancer pathogenesis (PTPRT, CDH24, PCLKC and SLC15A1). The other four had no previous association with cancer pathogenesis. They did have potential functions in metabolic pathways that suggested mechanisms by which they could act to promote cancer (KNDC1, GPR124, EB12, GRINC1B)

These genes are involved in pathways known to contribute to cancer pathogenesis, but before this study most would not have been candidates for targeted gene therapy. This analysis validated the approach of whole cancer genome sequencing in identifying somatic mutations and the importance of parallel sequencing of normal and tumor cell genomes.

In 2011, the genome of an exceptional bladder cancer patient whose tumor had been eliminated by the drug everolimus was sequenced, revealing mutations in two genes, TSC1 and NF2. The mutations disregulated mTOR, the protein inhibited by everolimus, allowing it to reproduce without limit. As a result, in 2015, the Exceptional Responders Initiative was created at the National Cancer Institute. The initiative allows such exceptional patients (who have responded positively for at least six months to a cancer drug that usually fails) to have their genomes sequenced to identify the relevant mutations. Once identified, other patients could be screened for those mutations and then be given the drug. In 2016 To that end, a nationwide cancer drug trial began in 2015, involving up to twenty-four hundred centers. Patients with appropriate mutations are matched with one of more than forty drugs.

In 2014 the Center for Molecular Oncology rolled out the MSK-IMPACT test, a screening tool that looks for mutations in 341 cancer-associated genes. By 2015 more than five thousand patients had been screened. Patients with appropriate mutations are eligible to enroll in clinical trials that provide targeted therapy.

Technologies

Current technologies being used in Oncogenomics.

Genomics technologies include:

Genome sequencing

  • DNA sequencing: Pyrosequencing-based sequencers offer a relatively low-cost method to generate sequence data.
  • Array Comparative Genome Hybridization: This technique measures the DNA copy number differences between normal and cancer genomes. It uses the fluorescence intensity from fluorescent-labeled samples, which are hybridized to known probes on a microarray.
  • Representational oligonucleotide microarray analysis: Detects copy number variation using amplified restriction-digested genomic fragments that are hybridized to human oligonucleotides, achieving a resolution between 30 and 35 kbit/s.
  • Digital Karyotyping: Detects copy number variation using genomics tags obtained via restriction enzyme digests. These tags are then linked to into ditags, concatenated, cloned, sequenced and mapped back to the reference genome to evaluate tag density.
  • Bacterial Artificial Chromosome (BAC)-end sequencing (end-sequence profiling): Identifies chromosomal breakpoints by generating a BAC library from a cancer genome and sequencing their ends. The BAC clones that contain chromosome aberrations have end sequences that do not map to a similar region of the reference genome, thus identifying a chromosomal breakpoint.

Transcriptomes

  • Microarrays: Assess transcript abundance. Useful in classification, prognosis, raise the possibility of differential treatment approaches and aid identification of mutations in the proteins' coding regions. The relative abundance of alternative transcripts has become an important feature of cancer research. Particular alternative transcript forms correlate with specific cancer types.
  • RNA-Seq

Bioinformatics and functional analysis of oncogenes

Bioinformatics technologies allow the statistical analysis of genomic data. The functional characteristics of oncogenes has yet to be established. Potential functions include their transformational capabilities relating to tumour formation and specific roles at each stage of cancer development.

After the detection of somatic cancer mutations across a cohort of cancer samples, bioinformatic computational analyses can be carried out to identify likely functional and likely driver mutations. There are three main approaches routinely used for this identification: mapping mutations, assessing the effect of mutation of the function of a protein or a regulatory element and finding signs of positive selection across a cohort of tumors. The approaches are not necessarily sequential however, there are important relationships of precedence between elements from the different approaches. Different tools are used at each step.

Operomics

Operomics aims to integrate genomics, transcriptomics and proteomics to understand the molecular mechanisms that underlie the cancer development.

Comparative oncogenomics

Comparative oncogenomics uses cross-species comparisons to identify oncogenes. This research involves studying cancer genomes, transcriptomes and proteomes in model organisms such as mice, identifying potential oncogenes and referring back to human cancer samples to see whether homologues of these oncogenes are important in causing human cancers. Genetic alterations in mouse models are similar to those found in human cancers. These models are generated by methods including retroviral insertion mutagenesis or graft transplantation of cancerous cells.

Source of cancer driver mutations, cancer mutagenesis

Mutations provide the raw material for natural selection in evolution and can be caused by errors of DNA replication, the action of exogenous mutagens or endogenous DNA damage. The machinery of replication and genome maintenance can be damaged by mutations, or altered by physiological conditions and differential levels of expression in cancer.

As pointed out by Gao et al., the stability and integrity of the human genome are maintained by the DNA-damage response (DDR) system. Un-repaired DNA damage is a major cause of mutations that drive carcinogenesis. If DNA repair is deficient, DNA damage tends to accumulate. Such excess DNA damage can increase mutational errors during DNA replication due to error-prone translesion synthesis. Excess DNA damage can also increase epigenetic alterations due to errors during DNA repair. Such mutations and epigenetic alterations can give rise to cancer. DDR genes are often repressed in human cancer by epigenetic mechanisms. Such repression may involve DNA methylation of promoter regions or repression of DDR genes by a microRNA. Epigenetic repression of DDR genes occurs more frequently than gene mutation in many types of cancer (see Cancer epigenetics). Thus, epigenetic repression often plays a more important role than mutation in reducing expression of DDR genes. This reduced expression of DDR genes is likely an important driver of carcinogenesis.

Nucleotide sequence context influences mutation probability and analysis of mutational (mutable) DNA motifs can be essential for understanding the mechanisms of mutagenesis in cancer. Such motifs represent the fingerprints of interactions between DNA and mutagens, between DNA and repair/replication/modification enzymes. Examples of motifs are the AID motif WRCY/RGYW (W = A or T, R = purine and Y = pyrimidine) with C to T/G/A mutations, and error-prone DNA pol η attributed AID-related mutations (A to G/C/G) in WA/TW motifs.

Another (agnostic) way to analyze the observed mutational spectra and DNA sequence context of mutations in tumors involves pooling all mutations of different types and contexts from cancer samples into a discrete distribution. If multiple cancer samples are available, their context-dependent mutations can be represented in the form of a nonnegative matrix. This matrix can be further decomposed into components (mutational signatures) which ideally should describe individual mutagenic factors. Several computational methods have been proposed for solving this decomposition problem. The first implementation of Non-negative Matrix Factorization (NMF) method is available in Sanger Institute Mutational Signature Framework in the form of a MATLAB package. On the other hand, if mutations from a single tumor sample are only available, the DeconstructSigs R package and MutaGene server may provide the identification of contributions of different mutational signatures for a single tumor sample. In addition, MutaGene server provides mutagen or cancer-specific mutational background models and signatures that can be applied to calculate expected DNA and protein site mutability to decouple relative contributions of mutagenesis and selection in carcinogenesis.

Synthetic lethality

Synthetic lethality arises when a combination of deficiencies in the expression of two or more genes leads to cell death, whereas a deficiency in only one of these genes does not. The deficiencies can arise through mutations, epigenetic alterations or inhibitors of one of the genes.

The therapeutic potential of synthetic lethality as an efficacious anti-cancer strategy is continually improving. Recently, the applicability of synthetic lethality to targeted cancer therapy has heightened due to the recent work of scientists including Ronald A. DePinho and colleagues, in what is termed 'collateral lethality'. Muller et al. found that passenger genes, with chromosomal proximity to tumor suppressor genes, are collaterally deleted in some cancers. Thus, the identification of collaterally deleted redundant genes carrying out an essential cellular function may be the untapped reservoir for then pursuing a synthetic lethality approach. Collateral lethality therefore holds great potential in identification of novel and selective therapeutic targets in oncology. In 2012, Muller et al. identified that homozygous deletion of redundant-essential glycolytic ENO1 gene in human glioblastoma (GBM) is the consequence of proximity to 1p36 tumor suppressor locus deletions and may hold potential for a synthetic lethality approach to GBM inhibition. ENO1 is one of three homologous genes (ENO2, ENO3) that encodes the mammalian alpha-enolase enzyme. ENO2, which encodes enolase 2, is mostly expressed in neural tissues, leading to the postulation that in ENO1-deleted GBM, ENO2 may be the ideal target as the redundant homologue of ENO1. Muller found that both genetic and pharmacological ENO2 inhibition in GBM cells with homozygous ENO1 deletion elicits a synthetic lethality outcome by selective killing of GBM cells. In 2016, Muller and colleagues discovered antibiotic SF2312 as a highly potent nanomolar-range enolase inhibitor which preferentially inhibits glioma cell proliferation and glycolytic flux in ENO1-deleted cells. SF2312 was shown to be more efficacious than pan-enolase inhibitor PhAH and have more specificity for ENO2 inhibition over ENO1. Subsequent work by the same team showed that the same approach could be applied to pancreatic cancer, whereby homozygously deleted SMAD4 results in the collateral deletion of mitochondrial malic enzyme 2 (ME2), an oxidative decarboxylase essential for redox homeostasis. Dey et al. show that ME2 genomic deletion in pancreatic ductal adenocarcinoma cells results in high endogenous reactive oxygen species, consistent with KRAS-driven pancreatic cancer, and essentially primes ME2-null cells for synthetic lethality by depletion of redundant NAD(P)+-dependent isoform ME3. The effects of ME3 depletion were found to be mediated by inhibition of de novo nucleotide synthesis resulting from AMPK activation and mitochondrial ROS-mediated apoptosis. Meanwhile, Oike et al. demonstrated the generalizability of the concept by targeting redundant essential-genes in process other than metabolism, namely the SMARCA4 and SMARCA2 subunits in the chromatin-remodeling SWI/SNF complex.

Some oncogenes are essential for survival of all cells (not only cancer cells). Thus, drugs that knock out these oncogenes (and thereby kill cancer cells) may also damage normal cells, inducing significant illness. However, other genes may be essential to cancer cells but not to healthy cells.

Treatments based on the principle of synthetic lethality have prolonged the survival of cancer patients, and show promise for future advances in reversal of carcinogenesis. A major type of synthetic lethality operates on the DNA repair defect that often initiates a cancer, and is still present in the tumor cells. Some examples are given here.

BRCA1 or BRCA2 expression is deficient in a majority of high-grade breast and ovarian cancers, usually due to epigenetic methylation of its promoter or epigenetic repression by an over-expressed microRNA (see articles BRCA1 and BRCA2). BRCA1 and BRCA2 are important components of the major pathway for homologous recombinational repair of double-strand breaks. If one or the other is deficient, it increases the risk of cancer, especially breast or ovarian cancer. A back-up DNA repair pathway, for some of the damages usually repaired by BRCA1 and BRCA2, depends on PARP1. Thus, many ovarian cancers respond to an FDA-approved treatment with a PARP inhibitor, causing synthetic lethality to cancer cells deficient in BRCA1 or BRCA2. This treatment is also being evaluated for breast cancer and numerous other cancers in Phase III clinical trials in 2016.

There are two pathways for homologous recombinational repair of double-strand breaks. The major pathway depends on BRCA1, PALB2 and BRCA2 while an alternative pathway depends on RAD52. Pre-clinical studies, involving epigenetically reduced or mutated BRCA-deficient cells (in culture or injected into mice), show that inhibition of RAD52 is synthetically lethal with BRCA-deficiency.

Mutations in genes employed in DNA mismatch repair (MMR) cause a high mutation rate. In tumors, such frequent subsequent mutations often generate “non-self” immunogenic antigens. A human Phase II clinical trial, with 41 patients, evaluated one synthetic lethal approach for tumors with or without MMR defects. The product of gene PD-1 ordinarily represses cytotoxic immune responses. Inhibition of this gene allows a greater immune response. When cancer patients with a defect in MMR in their tumors were exposed to an inhibitor of PD-1, 67% - 78% of patients experienced immune-related progression-free survival. In contrast, for patients without defective MMR, addition of PD-1 inhibitor generated only 11% of patients with immune-related progression-free survival. Thus inhibition of PD-1 is primarily synthetically lethal with MMR defects.

ARID1A, a chromatin modifier, is required for non-homologous end joining, a major pathway that repairs double-strand breaks in DNA, and also has transcription regulatory roles. ARID1A mutations are one of the 12 most common carcinogenic mutations. Mutation or epigenetically decreased expression of ARID1A has been found in 17 types of cancer. Pre-clinical studies in cells and in mice show that synthetic lethality for ARID1A deficiency occurs by either inhibition of the methyltransferase activity of EZH2, or with addition of the kinase inhibitor dasatinib.

Another approach is to individually knock out each gene in a genome and observe the effect on normal and cancerous cells. If the knockout of an otherwise nonessential gene has little or no effect on healthy cells, but is lethal to cancerous cells containing a mutated oncogene, then the system-wide suppression of the suppressed gene can destroy cancerous cells while leaving healthy ones relatively undamaged. The technique was used to identify PARP-1 inhibitors to treat BRCA1/BRCA2-associated cancers. In this case, the combined presence of PARP-1 inhibition and of the cancer-associated mutations in BRCA genes is lethal only to the cancerous cells.

Databases for cancer research

The Cancer Genome Project is an initiative to map out all somatic mutations in cancer. The project systematically sequences the exons and flanking splice junctions of the genomes of primary tumors and cancerous cell lines. COSMIC software displays the data generated from these experiments. As of February 2008, the CGP had identified 4,746 genes and 2,985 mutations in 1,848 tumours.

The Cancer Genome Anatomy Project includes information of research on cancer genomes, transcriptomes and proteomes.

Progenetix is an oncogenomic reference database, presenting cytogenetic and molecular-cytogenetic tumor data.

Oncomine has compiled data from cancer transcriptome profiles.

The integrative oncogenomics database IntOGen and the Gitools datasets integrate multidimensional human oncogenomic data classified by tumor type. The first version of IntOGen focused on the role of deregulated gene expression and CNV in cancer. A later version emphasized mutational cancer driver genes across 28 tumor types. All releases of IntOGen data are made available at the IntOGen database.

The International Cancer Genome Consortium is the biggest project to collect human cancer genome data. The data is accessible through the ICGC website. The BioExpress® Oncology Suite contains gene expression data from primary, metastatic and benign tumor samples and normal samples, including matched adjacent controls. The suite includes hematological malignancy samples for many well-known cancers.

Specific databases for model animals include the Retrovirus Tagged Cancer Gene Database (RTCGD) that compiled research on retroviral and transposon insertional mutagenesis in mouse tumors.

Gene families

Mutational analysis of entire gene families revealed that genes of the same family have similar functions, as predicted by similar coding sequences and protein domains. Two such classes are the kinase family, involved in adding phosphate groups to proteins and the phosphatase family, involved with removing phosphate groups from proteins. These families were first examined because of their apparent role in transducing cellular signals of cell growth or death. In particular, more than 50% of colorectal cancers carry a mutation in a kinase or phosphatase gene. Phosphatidylinositold 3-kinases (PIK3CA) gene encodes for lipid kinases that commonly contain mutations in colorectal, breast, gastric, lung and various other cancers. Drug therapies can inhibit PIK3CA. Another example is the BRAF gene, one of the first to be implicated in melanomas. BRAF encodes a serine/threonine kinase that is involved in the RAS-RAF-MAPK growth signaling pathway. Mutations in BRAF cause constitutive phosphorylation and activity in 59% of melanomas. Before BRAF, the genetic mechanism of melanoma development was unknown and therefore prognosis for patients was poor.

Mitochondrial DNA

Mitochondrial DNA (mtDNA) mutations are linked the formation of tumors. Four types of mtDNA mutations have been identified:

Point mutations

Point mutations have been observed in the coding and non-coding region of the mtDNA contained in cancer cells. In individuals with bladder, head/neck and lung cancers, the point mutations within the coding region show signs of resembling each other. This suggests that when a healthy cell transforms into a tumor cell (a neoplastic transformation) the mitochondria seem to become homogenous. Abundant point mutations located within the non-coding region, D-loop, of the cancerous mitochondria suggest that mutations within this region might be an important characteristic in some cancers.

Deletions

This type of mutation is sporadically detected due to its small size ( < 1kb). The appearance of certain specific mtDNA mutations (264-bp deletion and 66-bp deletion in the complex 1 subunit gene ND1) in multiple types of cancer provide some evidence that small mtDNA deletions might appear at the beginning of tumorigenesis. It also suggests that the amount of mitochondria containing these deletions increases as the tumor progresses. An exception is a relatively large deletion that appears in many cancers (known as the "common deletion"), but more mtDNA large scale deletions have been found in normal cells compared to tumor cells. This may be due to a seemingly adaptive process of tumor cells to eliminate any mitochondria that contain these large scale deletions (the "common deletion" is > 4kb).

Insertions

Two small mtDNA insertions of ~260 and ~520 bp can be present in breast cancer, gastric cancer, hepatocellular carcinoma (HCC) and colon cancer and in normal cells. No correlation between these insertions and cancer are established.

Copy number mutations

The characterization of mtDNA via real-time polymerase chain reaction assays shows the presence of quantitative alteration of mtDNA copy number in many cancers. Increase in copy number is expected to occur because of oxidative stress. On the other hand, decrease is thought to be caused by somatic point mutations in the replication origin site of the H-strand and/or the D310 homopolymeric c-stretch in the D-loop region, mutations in the p53 (tumor suppressor gene) mediated pathway and/or inefficient enzyme activity due to POLG mutations. Any increase/decrease in copy number then remains constant within tumor cells. The fact that the amount of mtDNA is constant in tumor cells suggests that the amount of mtDNA is controlled by a much more complicated system in tumor cells, rather than simply altered as a consequence of abnormal cell proliferation. The role of mtDNA content in human cancers apparently varies for particular tumor types or sites.

Mutations in mitochondrial DNA in various cancers
Cancer Type Location of Point mutations Nucleotide Position of Deletions Increase of mtDNA copy # Decrease of mtDNA copy #
D-Loop mRNAs tRNAs rRNAs
Bladder X X
X 15,642-15,662

Breast X X X X 8470-13,447 and 8482-13459
X
Head and neck X X X X 8470-13,447 and 8482-13459 X
Oral X X

8470-13,447 and 8482-13459

Hepatocellular carcinoma (HCC) X X X X 306-556 and 3894-3960
X
Esophageal X X
X 8470-13,447 and 8482-13459 X
Gastric  X X X
298-348
X
Prostate X

X 8470-13,447 and 8482-13459 X

57.7% (500/867) contained somatic point putations and of the 1172 mutations surveyed 37.8% (443/1127) were located in the D-loop control region, 13.1% (154/1172) were located in the tRNA or rRNA genes and 49.1% (575/1127) were found in the mRNA genes needed for producing complexes required for mitochondrial respiration.

Diagnostic applications

Some anticancer drugs target mtDNA and have shown positive results in killing tumor cells. Research has used mitochondrial mutations as biomarkers for cancer cell therapy. It is easier to target mutation within mitochondrial DNA versus nuclear DNA because the mitochondrial genome is much smaller and easier to screen for specific mutations. MtDNA content alterations found in blood samples might be able to serve as a screening marker for predicting future cancer susceptibility as well as tracking malignant tumor progression. Along with these potential helpful characteristics of mtDNA, it is not under the control of the cell cycle and is important for maintaining ATP generation and mitochondrial homeostasis. These characteristics make targeting mtDNA a practical therapeutic strategy.

Cancer biomarkers

Several biomarkers can be useful in cancer staging, prognosis and treatment. They can range from single-nucleotide polymorphisms (SNPs), chromosomal aberrations, changes in DNA copy number, microsatellite instability, promoter region methylation, or even high or low protein levels.

Cancer immunology

From Wikipedia, the free encyclopedia
 
Tumor-associated immune cells in the tumor microenvironment (TME) of breast cancer models

Cancer immunology is an interdisciplinary branch of biology that is concerned with understanding the role of the immune system in the progression and development of cancer; the most well known application is cancer immunotherapy, which utilises the immune system as a treatment for cancer. Cancer immunosurveillance and immunoediting are based on protection against development of tumors in animal systems and (ii) identification of targets for immune recognition of human cancer.

Definition

Cancer immunology is an interdisciplinary branch of biology concerned with the role of the immune system in the progression and development of cancer; the most well known application is cancer immunotherapy, where the immune system is used to treat cancer. Cancer immunosurveillance is a theory formulated in 1957 by Burnet and Thomas, who proposed that lymphocytes act as sentinels in recognizing and eliminating continuously arising, nascent transformed cells. Cancer immunosurveillance appears to be an important host protection process that decreases cancer rates through inhibition of carcinogenesis and maintaining of regular cellular homeostasis. It has also been suggested that immunosurveillance primarily functions as a component of a more general process of cancer immunoediting.

Tumor antigens

Tumors may express tumor antigens that are recognized by the immune system and may induce an immune response. These tumor antigens are either TSA (Tumor-specific antigen) or TAA (Tumor-associated antigen).

Tumor-specific

Tumor-specific antigens (TSA) are antigens that only occur in tumor cells. TSAs can be products of oncoviruses like E6 and E7 proteins of human papillomavirus, occurring in cervical carcinoma, or EBNA-1 protein of EBV, occurring in Burkitt's lymphoma cells. Another example of TSAs are abnormal products of mutated oncogenes (e.g. Ras protein) and anti-oncogenes (e.g. p53).

Tumor-associated antigens

Tumor-associated antigens (TAA) are present in healthy cells, but for some reason they also occur in tumor cells. However, they differ in quantity, place or time period of expression. Oncofetal antigens are tumor-associated antigens expressed by embryonic cells and by tumors. Examples of oncofetal antigens are AFP (α-fetoprotein), produced by hepatocellular carcinoma, or CEA (carcinoembryonic antigen), occurring in ovarian and colon cancer. More tumor-associated antigens are HER2/neu, EGFR or MAGE-1.

Immunoediting

Cancer immunoediting is a process in which immune system interacts with tumor cells. It consists of three phases: elimination, equilibrium and escape. These phases are often referred to as "the three Es" of cancer immunoediting. Both adaptive and innate immune system participate in immunoediting.

In the elimination phase, the immune response leads to destruction of tumor cells and therefore to tumor suppression. However, some tumor cells may gain more mutations, change their characteristics and evade the immune system. These cells might enter the equilibrium phase, in which the immune system does not recognise all tumor cells, but at the same time the tumor does not grow. This condition may lead to the phase of escape, in which the tumor gains dominance over immune system, starts growing and establishes immunosuppressive environment.

As a consequence of immunoediting, tumor cell clones less responsive to the immune system gain dominance in the tumor through time, as the recognized cells are eliminated. This process may be considered akin to Darwinian evolution, where cells containing pro-oncogenic or immunosuppressive mutations survive to pass on their mutations to daughter cells, which may themselves mutate and undergo further selective pressure. This results in the tumor consisting of cells with decreased immunogenicity and can hardly be eliminated. This phenomenon was proven to happen as a result of immunotherapies of cancer patients.

Tumor evasion mechanisms

Multiple factors determine whether tumor cells will be eliminated by the immune system or will escape detection. During the elimination phase immune effector cells such as CTL's and NK cells with the help of dendritic and CD4+ T-cells are able to recognize and eliminate tumor cells.
  • CD8+ cytotoxic T cells are a fundamental element of anti-tumor immunity. Their TCR receptors recognise antigens presented by MHC class I and when bound, the Tc cell triggers its cytotoxic activity. MHC I are present on the surface of all nucleated cells. However, some cancer cells lower their MHC I expression and avoid being detected by the cytotoxic T cells. This can be done by mutation of MHC I gene or by lowering the sensitivity to IFN-γ (which influences the surface expression of MHC I). Tumor cells also have defects in antigen presentation pathway, what leads into down-regulation of tumor antigen presentations. Defects are for example in transporter associated with antigen processing (TAP) or tapasin. On the other hand, a complete loss of MHC I is a trigger for NK cells. Tumor cells therefore maintain a low expression of MHC I.
  • Another way to escape cytotoxic T cells is to stop expressing molecules essential for co-stimulation of cytotoxic T cells, such as CD80 or CD86.
  • Tumor cells express molecules to induce apoptosis or to inhibit T lymphocytes:
    • Expression of FasL on its surface, tumor cells may induce apoptosis of T lymphocytes by FasL-Fas interaction.
    • Expression of PD-L1 on the surface of tumor cells leads to suppression of T lymphocytes by PD1-PD-L1 interaction.
  • Tumor cells have gained resistance to effector mechanisms of NK and cytotoxic CD8+ T cell:

Tumor microenvironment

Immune checkpoints of immunosuppressive actions associated with breast cancer

Immunomodulation methods

Immune system is the key player in fighting cancer. As described above in mechanisms of tumor evasion, the tumor cells are modulating the immune response in their profit. It is possible to improve the immune response in order to boost the immunity against tumor cells.

  • monoclonal anti-CTLA4 and anti-PD-1 antibodies are called immune checkpoint inhibitors:
    • CTLA-4 is a receptor upregulated on the membrane of activated T lymphocytes, CTLA-4 CD80/86 interaction leads to switch off of T lymphocytes. By blocking this interaction with monoclonal anti CTLA-4 antibody we can increase the immune response. An example of approved drug is ipilimumab.
    • PD-1 is also an upregulated receptor on the surface of T lymphocytes after activation. Interaction PD-1 with PD-L1 leads to switching off or apoptosis. PD-L1 are molecules which can be produced by tumor cells. The monoclonal anti-PD-1 antibody is blocking this interaction thus leading to improvement of immune response in CD8+ T lymphocytes. An example of approved cancer drug is nivolumab.
    • Chimeric Antigen Receptor T cell
      • This CAR receptors are genetically engineered receptors with extracellular tumor specific binding sites and intracellular signalling domain that enables the T lymphocyte activation.
    • Cancer vaccine

Relationship to chemotherapy

Obeid et al. investigated how inducing immunogenic cancer cell death ought to become a priority of cancer chemotherapy. He reasoned, the immune system would be able to play a factor via a 'bystander effect' in eradicating chemotherapy-resistant cancer cells. However, extensive research is still needed on how the immune response is triggered against dying tumour cells.

Professionals in the field have hypothesized that 'apoptotic cell death is poorly immunogenic whereas necrotic cell death is truly immunogenic'. This is perhaps because cancer cells being eradicated via a necrotic cell death pathway induce an immune response by triggering dendritic cells to mature, due to inflammatory response stimulation. On the other hand, apoptosis is connected to slight alterations within the plasma membrane causing the dying cells to be attractive to phagocytic cells. However, numerous animal studies have shown the superiority of vaccination with apoptotic cells, compared to necrotic cells, in eliciting anti-tumor immune responses.

Thus Obeid et al. propose that the way in which cancer cells die during chemotherapy is vital. Anthracyclins produce a beneficial immunogenic environment. The researchers report that when killing cancer cells with this agent uptake and presentation by antigen presenting dendritic cells is encouraged, thus allowing a T-cell response which can shrink tumours. Therefore, activating tumour-killing T-cells is crucial for immunotherapy success.

However, advanced cancer patients with immunosuppression have left researchers in a dilemma as to how to activate their T-cells. The way the host dendritic cells react and uptake tumour antigens to present to CD4+ and CD8+ T-cells is the key to success of the treatment.

Childhood cancer

From Wikipedia, the free encyclopedia
 
Childhood cancer
Other namesPediatric cancer
Trying out hats to wear after chemotherapy - cropped.jpg
A girl trying out hats to wear after chemotherapy against a Wilms' tumor
SpecialtyPediatrics, oncology

Childhood cancer is cancer in a child. About 80% of childhood cancer cases can be successfully treated. thanks to the modern medical treatments and optimal patient care. However, only about 10% of children diagnosed with cancer reside in high-income countries where the necessary treatments and care is available. Childhood cancer represents only about 1% of all types of cancers diagnosed in children and adults. For this reason, childhood cancer is often ignored in control planning, contributing to the burden of missed opportunities for its diagnoses and management in countries that are low- and mid-income.

In the United States, an arbitrarily adopted standard of the ages used are 0–14 years inclusive, that is, up to 14 years 11.9 months of age. However, the definition of childhood cancer sometimes includes adolescents between 15 and 19 years old. Pediatric oncology is the branch of medicine concerned with the diagnosis and treatment of cancer in children.

Signs and symptoms

Leukemia

This is the most common type of cancer during childhood, and acute lymphoblastic leukemia (ALL) is most common in children. ALL usually develops in children between the ages of 1 and 10 (it could occur at any age). This type of cancer is more prevalent in males and in whites.

Signs & Symptoms:

Frequent delayed diagnosis (early symptoms are nonspecific)

Physical examination:

Important: It is recommended that a complete blood count is obtained (CBC) if any suspicious finding arise.

Central nervous system tumors

This is the second most common malignancy diagnosed during childhood.

Signs and Symptoms

  • Ataxia
  • Other gait disturbances (hydrocephalus due to aqueduct compression)
  • Cranial nerve abnormalities as a result of brainstem compression

Hodgkin's disease

The likelihood of developing Hodgkin's disease increases during childhood and it peaks in adolescence.  

Signs and Symptoms

  • Painless mass in the neck
  • Persistent cough secondary to a mediastinal mass
  • Less commonly: splenomegaly or enlarged axillary or inguinal lymph nodes
  • Intermittent fever
  • Drenching night sweats
  • Loss of greater than 10 percent of total body weight.
  • Anorexia
  • Fatigue
  • Pruritus
  • Persistent painless mass

Non-Hodgkin's lymphoma

This type of cancer is more common in older children, and it is less prevalent than Hodgkin's disease.

Signs and Symptoms

If abdomen is affected

If mediastinum is affected

If head and neck masses are affected

  • Palpable mass
  • Cranial nerve palsies
  • Nasal obstruction

Neuroblastoma

This cancer is an extracranial solid tumor commonly diagnosed in childhood.  

Signs and Symptoms

  • Dysfunction of the location of the primary tumor
  • Anorexia
  • Abdominal pain
  • Distention.

Wilms' tumor

This malignancy presents as an abdominal mass in a child.

Signs and Symptoms

Malignancies of the musculoskeletal system

A tumor that arises in the musculoskeletal system often presents as a mass, a painful extremity or, occasionally, a pathologic fracture.

Signs and Symptoms

  • Pain awakens a child at night
  • Significant extremity dysfunction (when trauma is not involved)

Genetic syndromes associated with cancer

The cause of cancer is not yet well understood. Several chromosomal disorders and constitutional syndromes are associated with it.

Learning problems

Children with cancer are at risk for developing various cognitive or learning problems. These difficulties may be related to brain injury stemming from the cancer itself, such as a brain tumor or central nervous system metastasis or from side effects of cancer treatments such as chemotherapy and radiation therapy. Studies have shown that chemo and radiation therapies may damage brain white matter and disrupt brain activity.

This cognitive problem is known as post-chemotherapy cognitive impairment (PCCI) or "chemo brain." This term is commonly use by cancer survivors who describe having thinking and memory problems after cancer treatment. Researchers are unsure what exactly causes chemo brain, however, they say it is likely to be linked to either the cancer itself, the cancer treatment, or be an emotional reaction to both.

This cognitive impairment is commonly noticed a few years after a child endures cancer treatment. When a childhood cancer survivor goes back to school, they might experience lower test scores, problems with memory, attention, and behavior, as well as poor hand-eye coordination and slowed development over time. Children with cancer should be monitored and assessed for these neuropsychological deficits during and after treatment. Patients with brain tumors can have cognitive impairments before treatment  and radiation therapy is associated with increased risk of cognitive impairment. Parents can apply their children for special educational services at school if their cognitive learning disability affects their educational success.

Risk factors

Risk factors are any genetic or environmental exposure that increase the chances of developing a pathological condition. Some examples are age, family history, environmental factors, genetics, and economic status among others.

Demographic risk factors

  • Childhood cancer varies by age, sex, ethnicity, and race. Its incidence peaks in infancy with about 240 cases/million/year.
  • This rate decreases to 128 cases per million from 5–9 years of age, and it rises again to 220 cases/million.
  • Slight male dominance for most childhood cancers.

Environmental factors

  • High dose ionizing radiation and prior chemotherapy are accepted causes of childhood cancer, each raising risk several fold (4-7).

Genetic factors

Identified Cancer Predisposition Syndromes

  • Li-Fraumeni syndrome (TP53)
  • Hereditary breast or ovarian cancer (BRCA 12)
  • Colorectal cancer/polyposis syndromes
  • Familial retinoblastoma (RB1)
  • Familial and genetic factors are identified in 5-15% of childhood cancer cases. In <5-10% of cases, there are known environmental exposures and exogenous factors, such as prenatal exposure to tobacco, X-rays, or certain medications. For the remaining 75-90% of cases, however, the individual causes remain unknown. In most cases, as in carcinogenesis in general, the cancers are assumed to involve multiple risk factors and variables.

Aspects that make the risk factors of childhood cancer different from those seen in adult cancers include:

  • Different, and sometimes unique, exposures to environmental hazards. Children must often rely on adults to protect them from toxic environmental agents.
  • Immature physiological systems to clear or metabolize environmental substances
  • The growth and development of children in phases known as "developmental windows" result in certain "critical windows of vulnerability".

Also, a longer life expectancy in children avails for a longer time to manifest cancer processes with long latency periods, increasing the risk of developing some cancer types later in life.

Advanced parental age has been associated with increased risk of childhood cancer in the offspring. There are preventable causes of childhood malignancy, such as delivery overuse and misuse of ionizing radiation through computed tomography scans when the test is not indicated or when adult protocols are used.

Diagnosis

Types

The most common cancers in children are (childhood) leukemia (32%), brain tumors (18%), and lymphomas (11%). In 2005, 4.1 of every 100,000 young people under 20 years of age in the U.S. were diagnosed with leukemia, and 0.8 per 100,000 died from it. The number of new cases was highest among the 1–4 age group, but the number of deaths was highest among the 10–14 age group.

In 2005, 2.9 of every 100,000 people 0–19 years of age were found to have cancer of the brain or central nervous system, and 0.7 per 100,000 died from it. These cancers were found most often in children between 1 and 4 years of age, but the most deaths occurred among those aged 5–9. The main subtypes of brain and central nervous system tumors in children are: astrocytoma, brain stem glioma, craniopharyngioma, desmoplastic infantile ganglioglioma, ependymoma, high-grade glioma, medulloblastoma and atypical teratoid rhabdoid tumor.

Other, less common childhood cancer types are:

Medical specialties

Overall, treating childhood cancer requires a multidisciplinary team of doctors, nurses, social workers, therapists, and other members of the community. Here is a brief list of doctors that can treat childhood cancer:

  • Pediatric oncologist: These doctors specialize in treating childhood cancers.
  • Pediatric hematology-oncologist: These doctors specialize in treating blood diseases in children.
  • Pediatric surgeon: These doctors specialize in performing surgery on children.
  • Adolescent and young adult oncology (AYA): AYA is a branch of medicine that deals with the prevention, diagnosis, and treatment of cancer in adolescents and young adults, often defined as those aged 13–30. Studies have continuously shown that while pediatric cancer survival rates have gone up, the survival rate for adolescents and young adults has remained stagnant. Additionally, AYA helps patients with college concerns, fertility, and sense of aloneness. Studies have often shown that treating young adults with the same protocols used in pediatrics is more effective than adult-oriented treatments.

Other specialties that can assist in the treatment process include radiology, neurosurgery, orthopedic surgery, psychiatry, and endocrinology.

Treatment

Childhood cancer treatment is individualized and varies based on the severity & type of cancer. In general, treatment can include surgical resection, chemotherapy, radiation therapy, or immunotherapy.

Recent medical advances have improved our understanding of the genetic basis of childhood cancers. Treatment options are expanding, and precision medicine for childhood cancers is a rapidly growing area of research.

The side effects of chemotherapy can result in immediate and long-term treatment-related comorbidities. For children undergoing treatment for high-risk cancer, more than 80% experience life-threatening or fatal toxicity as a result of their treatment.

Psychosocial care of children with cancer is also important during the cancer journey, but the implementation of evidence-based interventions need to be further spread across pediatric cancer centers. In general, psychosocial care can include therapy with a psychologist or psychiatrist, referral to a social worker, or referral to a pastoral counselor. Family-centered psychosocial care is one approach that can be used to not only support the patient's psychosocial well-being but also support the parents and any caregivers of the patient.

Prognosis

With the advancement of new treatments for childhood cancer, 85% of individuals who had childhood cancer now survive 5 years or more. This is an increase from the mid-1970s where only 58% of children with childhood cancer survived 5 years or more. However, this survival rate is dependent on many factors such as the type of cancer, age of onset, location of the cancer, cancer stage, and if there is any genetic component to the cancer. Survival rate is also impacted by socioeconomic status and access to resources during treatment.

Since adult survivors of childhood cancer are living longer, these individuals may experience long-term complications that are associated with their cancer treatment. This can include problems with organ function, growth and development, neurocognitive function and academic achievement, and risk for additional cancers.

Premature heart disease is one example of a major long-term consequence seen in adult survivors of childhood cancer. These individuals are eight times more likely to die of heart disease than other people, and more than half of the children treated for cancer develop some type of cardiac abnormality, although this may be asymptomatic or too mild to qualify for a clinical diagnosis of heart disease.

Childhood cancer survivors are also at risk of sustaining adverse effects on the kidneys and the liver. Specific cancer treatments such as cisplatin, carboplatin, and radiotherapy are known to cause kidney damage. The risk of liver damage is increased in those who have had radiotherapy to the liver and in those with other risk factors, such as a higher body mass index or chronic viral hepatitis. Certain treatments and liver surgery may also increase the risk of adverse liver effects in childhood cancer survivors.

To help monitor for these long-term consequences, a set of guidelines have been created to facilitate long term follow up for childhood, adolescent, and young adult cancer survivors. This provides guidance for healthcare professionals on how to provide high quality follow-up care and appropriate monitoring. These guidelines also help healthcare providers collaborate with oncology specialists, in order to create recommendations specific to an individual patient.

Epidemiology

Epidemiology is the study of the distribution and determinants of disease frequency in the human population and the study of how to control health problems. Internationally, the greatest variation in childhood cancer incidence occurs when comparing high-income countries to low-income ones. This may result from differences in being able to diagnose cancer, differences in risk among different ethnic or racial population subgroups, as well as differences in risk factors. An example of differing risk factors is in cases of pediatric Burkitt lymphoma, a form of non-Hodgkin lymphoma that sickens 6 to 7 children out of every 100,000 annually in parts of sub-Saharan Africa, where it is associated with a history of infection by both Epstein-Barr virus and malaria. In industrialized countries, Burkitt lymphoma is not associated with these infectious diseases. Non-Hispanic white children often have a better chance of survival compared to other racial and ethnic groups. Where an individual lives is one of the biggest determinants of health in the world, as illness and healthcare options can vary by an individual's postal code.

United States

In the United States, cancer is the second most common cause of death among children between the ages of 1 and 14 years, exceeded only by unintentional injuries such as injuries sustained in a car wreck. More than 16 out of every 100,000 children and teens in the U.S. were diagnosed with cancer, and nearly 3 of every 100,000 died from the disease. In the United States in 2012, it was estimated that there was an incidence of 12,000 new cases, and 1,300 deaths, from cancer among children 0 to 14 years of age. Cancer is the second leading cause of death in males and fourth in women under the age of 20 in the United States. The survival rate of children with cancer has improved since the late 1960s which is due to improved treatment and public health measures. The estimated proportion surviving 5 years from diagnosis increased from 77.8 percent to 82.7 percent to 85.4 percent for those diagnosed in the 1990s, 2000s, and 2010–2016.

Statistics from the 2014 American Cancer Society report:

Ages birth to 14
Sex Incidence Mortality Observed Survival %
Boys 178.0 23.3 81.3
Girls 160.1 21.1 82.0
Ages 15 to 19
Sex Incidence Mortality Observed Survival %
Boys 237.7 34.5 80.0
Girls 235.5 24.7 85.4

Note: Incidence and mortality rates are per 1,000,000 and age-adjusted to the 2000 US standard population. Observed survival percentage is based on data from 2003-2009.

Sub-Saharan Africa

A large number of children in Africa live in low- and middle-income countries where there is limited access to prevention or treatment of cancer. The under-five mortality rate (U5MR), a robust indicator of child health, is at 109 per 1,000 live births. The proportion of childhood cancer is higher in Africa than in developed countries, at 4.8%. Kids with cancer are disadvantaged compared to kids in developed countries; therefore their statistic for childhood cancer is higher. In sub-Saharan Africa, 10% of children die before their 5th birthday, yet it is not due to cancer; communicable diseases such as malaria, cholera, and other infections are the leading cause of death. Children with cancer are often exposed to these preventable infections and diseases. Tumor registries only cover 11% of the African population, and there is a significant absence in death registration, making the mortality database unreliable. Overall, there is a lack of reliable data, as there is limited funding and many diseases are largely unknown to this population.

United Kingdom

Cancer in children is rare in the UK, with an average of 1,800 diagnoses every year but contributing to less than 1% of all cancer-related deaths. Age is not a confounding factor in mortality from the disease in the UK. From 2014 to 2016, approximately 230 children died from cancer, with brain/CNS cancers being the most commonly fatal type.

Foundations and fundraising

Part of the proceeds from the sale of yellow silage wrappings goes to childhood cancer research, Brastad, Sweden

Currently, there are various organizations whose main focus is fighting childhood cancer. Organizations focused on childhood cancer through cancer research and/or support programs include: Childhood Cancer Canada, Young Lives vs Cancer and the Children's Cancer and Leukaemia Group (in United Kingdom), Child Cancer Foundation (in New Zealand), Children's Cancer Recovery Foundation (in United States), American Childhood Cancer Organization (in United States), Childhood Cancer Support (Australia) and the Hayim Association (in Israel). Alex's Lemonade Stand Foundation allows people across the US to raise money for pediatric cancer research by organizing lemonade stands. The National Pediatric Cancer Foundation focuses on finding less toxic and more effective treatments for pediatric cancers. This foundation works with 24 different hospitals across the US in search of treatments effective in practice. Childhood Cancer International is the largest global pediatric cancer foundation. It focuses on early access to care for childhood cancers, focusing on patient support and patient advocacy.

According to estimates by experts in the field of pediatric cancer, by 2020, cancer will cost $158 million annually for both research and treatment which marks a 27% increase since 2010. Ways in which the foundations are helped by people include writing checks, collecting spare coins, bake/lemonade sales, donating portions of purchases from stores or restaurants, or Paid Time Off donations as well as auctions, bike rides, dance-a-thons. Additionally, many of the major foundations have donation buttons on their respective websites.

In addition to advancing research focusing on cancer, the foundations also offer support to families whose children are affected by the disease. The estimated total cost for one child with cancer (medical costs and lost parental wages) is $833,000. Organizations such as the National Children's Cancer Society and the Leukemia and Lymphoma Society can provide financial assistance for the costs associated with childhood cancer like medical care, home care, child care, and transportation.

Importance of family support

The emotional challenges that a parent may encounter can disrupt their child's treatment, parenting and support for the child who is ill and their siblings, and impact overall family stability. Therefore, having a support network during this time is important. Different foundations fund support groups within hospitals and online for parents and families to aid in the coping process. Targeted support for siblings of children with cancer is also warranted. Resources that account for family context, age, and gender can help siblings process cancer-related emotional reactions. These targeted resources help promote family activities and normal family functioning, while enhancing family adjustment over time.

The foundations for pediatric cancer all fall under the 501(c)3 designation which means that they are non-profit organizations that are tax-exempt. The "International Childhood Cancer Day" occurs annually on February 15.

Authorship of the Bible

From Wikipedia, the free encyclopedia ...