NANOGP8 is one of eleven NANOG pseudogenes in the human genome, specifically a processed pseudogene that retains a complete open reading frame despite its pseudogene classification. Unlike most pseudogenes, NANOGP8 is functional and expressed in certain contexts. The parent gene, NANOG, is located on chromosome 12p13 and functions as a transcription factor in embryonic stem cells, playing key roles in self-renewal and maintenance of pluripotency .
The primary differences between NANOGP8 and NANOG include:
Genomic location: NANOG is found on chromosome 12p13, while NANOGP8 is located on chromosome 15
Structure: NANOG contains introns, while NANOGP8 as a retrogene lacks introns
Expression pattern: NANOG is primarily expressed in embryonic stem cells, while NANOGP8 shows expression in various cancer cells and tissues
Evolutionary history: NANOGP8 is human-specific, having been inserted after human-chimpanzee divergence, making it the most recent of the NANOG pseudogenes
NANOGP8 has unique characteristics that distinguish it from other NANOG pseudogenes, including an Alu element in its 3'-UTR that is homologous to that of NANOG, and transcription factor binding sites in its SVA LTR that may promote its expression in cancer cells .
Differentiating NANOGP8 from NANOG and other pseudogenes requires specialized PCR strategies due to their high sequence similarity. The most effective approach involves careful primer design targeting unique regions, combined with size differentiation methods that exploit the structural differences between NANOG (containing introns) and NANOGP8 (lacking introns).
The following methodological strategy is recommended:
Primer design: Select primer pairs targeting sites unique to NANOG and/or NANOGP8 while excluding other NANOG pseudogenes. This requires identifying the few divergent nucleotide positions between these sequences .
Fragment size differentiation: Design primer pairs that co-amplify fragments from both NANOG and NANOGP8, but produce different fragment sizes due to the presence of introns in NANOG and their absence in NANOGP8 .
Verification steps: Use Primer-BLAST to confirm that primers will not amplify unintended targets from other NANOG pseudogenes .
A specific example from the research literature uses primer pair F2/R1 which amplifies fragments of different lengths:
1132 bp from NANOG
997 bp from NANOGP8
Similarly, primer pair F2/R2 produces:
These size differences are readily distinguishable through standard gel electrophoresis, allowing for reliable identification of the source sequence. Researchers should take special precautions during PCR setup to avoid contamination, including using clean rooms and DNA-free reagents .
NANOGP8 contains several key genetic variants that distinguish it from NANOG. These variants include both coding and non-coding changes, with three fixed differences and multiple polymorphic sites. Understanding these differences is crucial for accurate identification and functional studies.
The following table summarizes the key distinguishing variants between NANOG and NANOGP8:
Position | Variant | NANOG | NANOGP8 | Effect |
---|---|---|---|---|
c.144G>A | G>A | G | A | Synonymous |
c.759G>C | G>C | G | C | Gln253His |
c.*606T>G | T>G | T | G | 3' UTR |
c.47C>A | C>A | C | C/A | Ala16Glu |
c.190G>T | G>T | G | G/T | Asp64Tyr |
c.552A>T | A>T | A | A/T | Synonymous |
c.629C>T | C>T | C | C/T | Thr210Ile |
c.754A>C | A>C | A | A/C | Met252Leu |
The three variants shown in the first three rows (c.144G>A, c.759G>C, and c.*606T>G) are fixed differences between NANOG and NANOGP8, while the others represent polymorphic sites within NANOGP8 .
Additionally, NANOGP8 contains a 22-nucleotide deletion in the 3' UTR (c.*552-*573del) and variation in poly(T) mononucleotide repeats in the 3' UTR. These structural differences provide further markers for distinguishing between the two sequences .
Multiple lines of evidence firmly establish NANOGP8 as a human-specific genetic element that emerged after the human-chimpanzee divergence. This represents an important example of a functional retrogene that is uniquely human and may contribute to human-specific traits or disease susceptibilities.
The key evidence supporting NANOGP8's human specificity includes:
Absence in chimpanzee genome: Genomic sequence analysis reveals that NANOGP8 is completely absent from the chimpanzee genome, despite the high conservation of the parent NANOG gene across primate species .
Insertion timing: Comparative genomic analyses indicate that NANOGP8 was inserted into chromosome 15 after the human-chimpanzee divergence, making it the most recent of the NANOG pseudogenes .
Fixation in modern humans: Experimental evidence indicates that NANOGP8 is fixed in modern human populations, meaning all humans carry this genetic element, even though its parent allele shows polymorphism .
Sequence analysis: Alignment of human NANOG and NANOGP8 reference sequences with other NANOG pseudogenes and with NANOG genes from chimpanzee, orangutan, rhesus macaque, and gorilla confirms the human-specific nature of NANOGP8 .
This human specificity makes NANOGP8 particularly interesting from both evolutionary and medical perspectives, as it may contribute to human-specific characteristics including potentially higher genetic predisposition for certain cancers compared to other primates .
The evolution of NANOGP8 as a human-specific genetic element may contribute to increased cancer susceptibility in humans compared to other primates. Several mechanisms have been proposed to explain this relationship, supported by both genomic and functional evidence.
The potential links between NANOGP8 and human-specific disease susceptibility include:
Cancer cell expression: NANOGP8 has been found to be expressed in several cancer cell lines and in all cancer tissues tested, suggesting a potential role in tumorigenesis that would be unique to humans .
Transcriptional activation: NANOGP8 contains transcription factor binding sites in its SVA LTR (long terminal repeat) that may promote its expression specifically in cancer cells. This structural feature likely emerged after insertion into the human genome .
Proliferative effects: Experimental evidence shows that expression of NANOGP8 in NIH3T3 cells can promote cell proliferation, suggesting a potential oncogenic function .
Human cancer predisposition: Research suggests that the presence and expression of NANOGP8 may be a partial reason for the higher genetic predisposition for cancer in humans compared with other primates that lack this genetic element .
The human-specific nature of NANOGP8 represents an interesting case of how recent evolutionary changes in the human genome may influence disease susceptibility patterns. This retrogene appears to be fixed in modern human populations, suggesting potential selective advantages despite its association with cancer risk .
Determining the timing of NANOGP8 insertion into the human genome requires a multidisciplinary approach combining comparative genomics, molecular clock analysis, and population genetics. These complementary methodologies help establish that NANOGP8 was inserted after human-chimpanzee divergence.
The following methods are most effective for dating the insertion of NANOGP8:
Comparative genomic analysis: By examining the presence or absence of NANOGP8 across different primate species, researchers established that NANOGP8 is present in humans but absent in chimpanzees and other primates. This places the insertion event after the human-chimpanzee divergence, approximately 5-7 million years ago .
Sequence divergence analysis: Comparing the sequence of NANOGP8 to its parent gene NANOG allows calculation of the degree of divergence. By applying molecular clock principles (assuming a constant rate of neutral mutations), researchers can estimate the time since insertion.
Population genetic analysis: By analyzing NANOGP8 sequence data across diverse human populations, researchers determined that NANOGP8 is fixed in modern humans, suggesting the insertion occurred before the diversification of modern human populations .
Analysis of surrounding genomic regions: Examining the sequence and structure of DNA flanking the NANOGP8 insertion site can provide clues about the mechanism of insertion and potentially the timing.
Analysis of integration mechanism: The presence of features typical of retrotransposition, such as target site duplications or poly(A) tails, can confirm the mechanism of insertion and help establish the approximate timing.
These methods collectively provide strong evidence that NANOGP8 represents a relatively recent addition to the human genome that occurred after our evolutionary split from chimpanzees but before the diversification of modern human populations .
NANOGP8 has been detected in multiple cancer types, with consistent expression across various cancer tissues and cell lines. This widespread expression pattern suggests a potential universal role in tumorigenesis rather than a cancer type-specific function.
The evidence for NANOGP8 expression in cancer includes:
Cancer cell lines: Research has demonstrated NANOGP8 expression in several cancer cell lines across different tissue origins .
Cancer tissues: Studies have found that NANOGP8 is expressed in all cancer tissues tested, suggesting its potential relevance across multiple cancer types .
Complete coding sequence: The complete coding sequence of NANOGP8 has been cloned from cancer cells, confirming it maintains a functional open reading frame highly homologous to that of NANOG .
Protein expression: NANOGP8 protein expression has been detected using anti-NANOG antibodies in both recombinant E. coli systems and some cancer cell lines, confirming translation of the mRNA into protein .
This expression pattern is particularly notable because NANOGP8 was previously considered unlikely to be expressed, as it had not been identified in any expressed sequence tags (ESTs) prior to these cancer-focused investigations . The discovery of NANOGP8 expression specifically in cancer contexts suggests it may have acquired unique regulatory elements that activate its expression during tumorigenesis, potentially through the transcription factor binding sites in its SVA LTR .
NANOGP8 appears to contribute to tumorigenesis through multiple mechanisms, functioning as a potential retro-oncogene with functional impacts on cell proliferation and cancer development. Several lines of evidence support specific pathways through which NANOGP8 may promote cancer progression.
The primary mechanisms through which NANOGP8 may contribute to tumorigenesis include:
Cell proliferation promotion: Experimental evidence demonstrates that expression of NANOGP8 in NIH3T3 cells promotes cell proliferation, indicating a direct functional effect on a key cancer hallmark .
Stem cell-like properties: Given that the parent gene NANOG functions as a transcription factor involved in self-renewal and pluripotency maintenance in embryonic stem cells, NANOGP8 may confer stem cell-like properties to cancer cells. This could potentially contribute to cancer stem cell formation or maintenance .
Unique regulatory elements: NANOGP8 contains transcription factor binding sites in its SVA LTR that may promote its specific expression in cancer cells, providing a mechanism for its cancer-associated activation .
Protein functionality: NANOGP8 protein has been detected using anti-NANOG antibody in cancer cell lines, suggesting it produces a functional protein that may retain some of the transcriptional regulatory functions of NANOG .
Human-specific cancer risk: The presence of NANOGP8 has been proposed as a partial explanation for higher genetic predisposition for cancer in humans compared with other primates, suggesting it may confer unique cancer susceptibility .
These findings collectively position NANOGP8 as a retro-oncogene with potential regulatory functions in cancer development. Its expression across multiple cancer types suggests it may play important roles in general tumorigenesis mechanisms rather than cancer-specific processes .
Studying NANOGP8 function in cancer cells requires specialized approaches that can distinguish it from the parent NANOG gene and evaluate its specific contributions to cancer phenotypes. Several complementary experimental strategies have proven effective.
The most productive experimental approaches include:
Specific gene expression analysis:
Protein expression detection:
Functional interrogation:
Cancer phenotype assessment:
Cell proliferation assays
Colony formation assays
Invasion and migration assays
In vivo tumorigenicity studies in mouse models
Molecular interaction studies:
ChIP-seq to identify genomic binding sites
RNA-seq to assess transcriptional effects
Co-immunoprecipitation to identify protein interaction partners
These approaches must carefully address the challenge of distinguishing NANOGP8 from NANOG and other pseudogenes. Proper experimental controls should include verification of specificity through sequencing and the use of multiple detection methods to confirm results .
NANOGP8 exhibits several polymorphisms across human populations, despite being fixed as a genetic element in modern humans. These variations provide insights into its evolutionary history and potential functional differences.
The key polymorphisms identified in NANOGP8 include:
Coding region variants:
5' UTR variants:
3' UTR variants:
Poly(T) repeat variations:
Analysis of these polymorphisms across diverse human populations (including samples from the SNP500Cancer panel and Africans South of the Sahara panel) shows that while NANOGP8 itself is fixed in modern humans, its sequence contains considerable variation . This pattern suggests that NANOGP8 was inserted into the human genome before the diversification of modern human populations but recently enough that significant polymorphism remains.
The functional consequences of these polymorphisms remain largely unexplored, but the amino acid changes could potentially affect protein function, while the UTR variations might influence expression levels or post-transcriptional regulation .
Differentiating between NANOGP8 and NANOG expression presents significant technical challenges due to their high sequence similarity. Researchers need specialized strategies that exploit the structural and sequence differences between these genes.
The following methodological approaches are most effective:
PCR-based differentiation:
Sequence-specific detection:
Design TaqMan probes or molecular beacons targeting distinguishing sequences
Use allele-specific PCR primers that will only extend on the correct template
Implement high-resolution melt curve analysis to differentiate amplicons based on sequence composition
Restriction fragment length polymorphism (RFLP):
Identify restriction enzyme recognition sites present in only one of the two sequences
Digest PCR products and analyze fragment patterns
RNA sequencing analysis:
Look for reads spanning splice junctions (unique to NANOG)
Identify variant-specific reads at positions known to differ between NANOG and NANOGP8
Protein-level differentiation:
Use mass spectrometry to identify peptides containing amino acid differences
Generate antibodies specific to unique epitopes in NANOGP8
When implementing these approaches, researchers should follow these best practices:
Always sequence verify representative samples to confirm specificity
Include positive controls for both NANOG and NANOGP8
Use multiple independent methods to corroborate findings
Take special precautions to avoid contamination, including the use of clean rooms and DNA-free reagents
An example primer strategy from the literature uses the primer pair F3/R1 to specifically amplify a 1029 bp fragment from NANOG and the primer pair F4/R1 to specifically amplify a 990 bp fragment from NANOGP8 .
Sequencing and analyzing NANOGP8 presents several technical challenges that require specialized approaches to overcome. These difficulties arise from its high similarity to NANOG and other pseudogenes, as well as structural features of the sequence itself.
The primary technical challenges include:
Sequence similarity issues:
NANOGP8 shares exceptionally high sequence homology with NANOG and other NANOG pseudogenes
This limits the identification of unique primer binding sites as most potential sites could result in co-amplification of unintended fragments
Many potential amplification products would be identical in size, further complicating identification
Primer design constraints:
Researchers must carefully select primer pairs that target the few sites unique to NANOGP8
In some cases, primers must be designed to co-amplify fragments from both NANOG and NANOGP8, relying on size differences for discrimination
Extensive in silico verification with tools like Primer-BLAST is essential to ensure specificity
Contamination risks:
Sequence verification requirements:
Analysis complexities:
A successful approach documented in the literature involves amplifying a 1681 bp fragment comprising 78% of the complete NANOGP8 sequence using primer pair F1/R1, followed by cloning, plasmid preparation, and sequencing with both standard M13 primers and internal primers F4 and R5 .
Selecting appropriate cell models is crucial for meaningful investigation of NANOGP8 function. The ideal models should reflect relevant biological contexts while enabling specific manipulation and assessment of NANOGP8 activity.
The following cell models are particularly valuable for NANOGP8 research:
Cancer cell lines with confirmed NANOGP8 expression:
NIH3T3 cells for overexpression studies:
Embryonic stem cell models:
Given the role of the parent NANOG gene in stem cell biology, ES cells allow investigation of potential stem cell-related functions
Comparative studies between NANOG and NANOGP8 effects are possible
May reveal insights into cancer stem cell properties
CRISPR-engineered isogenic cell lines:
Creation of matched cell lines with and without NANOGP8 expression
Enables precise assessment of NANOGP8's contribution to cellular phenotypes
Controls for genetic background variables
Patient-derived cancer models:
Primary cancer cells from patients
Patient-derived xenografts (PDX)
Organoids that maintain tissue architecture
Each model system offers different advantages, and researchers should consider using multiple complementary models to build a comprehensive understanding of NANOGP8 function. When designing experiments, specific attention should be paid to methods that can distinguish NANOGP8 from NANOG effects, including the use of NANOGP8-specific targeting strategies and appropriate controls .
Measuring NANOGP8's impact on cell proliferation requires robust methodologies that can attribute observed effects specifically to NANOGP8 rather than NANOG or other factors. A comprehensive assessment approach using multiple complementary techniques is recommended.
The following methodological framework is most effective:
Genetic manipulation approaches:
Proliferation assay battery:
Real-time cell analysis systems (e.g., xCELLigence) for continuous monitoring
MTT/MTS/WST-1 assays for metabolic activity assessment
BrdU incorporation to measure DNA synthesis
Ki-67 immunostaining to identify actively dividing cells
Colony formation assays for long-term proliferative capacity
Cell cycle analysis using flow cytometry
Experimental design considerations:
Controls and validation:
Empty vector controls for overexpression studies
Non-targeting control siRNAs for knockdown experiments
Quantification of NANOGP8 expression levels by qRT-PCR and western blotting
Verification that NANOG expression remains unchanged
Rescue experiments to confirm phenotype is due to NANOGP8
In vivo validation:
Xenograft models to assess effects on tumor growth
Patient-derived samples correlation studies
This comprehensive approach builds upon previous research showing that NANOGP8 expression in NIH3T3 cells promotes cell proliferation, but extends it with more rigorous controls and multiple measurement techniques to establish causality and mechanism .
Studying interactions between NANOGP8 and other oncogenic pathways requires integrated approaches that can capture complex regulatory relationships and functional interactions. These methodologies should address both direct physical interactions and indirect pathway effects.
The following approaches are most effective for investigating these interactions:
Transcriptomic analysis:
RNA-seq comparing cells with and without NANOGP8 expression
Analysis of differentially expressed genes to identify affected pathways
GSEA (Gene Set Enrichment Analysis) to determine pathway enrichment
Time-course experiments to capture dynamic changes following NANOGP8 induction
Protein interaction studies:
Co-immunoprecipitation to identify direct protein binding partners
Proximity ligation assay (PLA) to detect protein-protein interactions in situ
BioID or APEX2 proximity labeling to identify proteins in the same complex
Yeast two-hybrid screening for systematic interaction mapping
Chromatin studies:
ChIP-seq to identify genomic binding sites of NANOGP8 protein
ATAC-seq to assess changes in chromatin accessibility
ChIP-seq for histone modifications to understand epigenetic effects
CUT&RUN or CUT&Tag for higher resolution protein-DNA interaction mapping
Functional genomics:
CRISPR screens to identify synthetic lethal interactions with NANOGP8
Double knockdown/knockout experiments to test specific pathway interactions
Small molecule inhibitor panels to identify pathway vulnerabilities
Systems biology integration:
Network analysis to position NANOGP8 within oncogenic signaling networks
Computational modeling of pathway interactions
Integration of multi-omics data (transcriptome, proteome, epigenome)
Validation in multiple models:
Testing identified interactions across different cancer types
Patient-derived models to confirm clinical relevance
In vivo models to validate in physiological context
These approaches should focus particularly on pathways related to stemness and self-renewal, given the function of the parent NANOG gene, as well as established oncogenic pathways including cell cycle regulation, apoptosis resistance, and metabolic reprogramming. The potential role of NANOGP8 as a human-specific cancer driver makes understanding its pathway interactions especially important for developing targeted therapeutic approaches that could exploit these unique vulnerabilities .
Targeting NANOGP8 represents a potentially valuable therapeutic strategy given its human-specific nature and apparent role in cancer progression. Several approaches show particular promise for translating NANOGP8 research into cancer therapeutics.
The most promising therapeutic approaches include:
RNA interference-based therapeutics:
siRNA or antisense oligonucleotides targeting NANOGP8-specific sequences
Delivery using lipid nanoparticles or other targeted delivery systems
Advantage of high specificity due to ability to target unique sequence regions
CRISPR-based therapies:
Guide RNAs targeting NANOGP8-specific sequences
Base editing approaches to introduce inactivating mutations
Potential for permanent inactivation compared to transient RNA interference
Protein-targeted approaches:
Development of small molecule inhibitors of NANOGP8 protein function
Peptide inhibitors that disrupt key protein-protein interactions
Degraders (PROTACs) that could selectively target NANOGP8 protein for degradation
Immunotherapeutic strategies:
Development of antibodies targeting NANOGP8-specific epitopes
CAR-T cell approaches targeting cancer cells expressing NANOGP8
Cancer vaccines eliciting immune responses against NANOGP8-expressing cells
Synthetic lethality exploitation:
Identification of genes/pathways that become essential in the context of NANOGP8 expression
Development of inhibitors targeting these synthetic lethal partners
Combination therapies exploiting NANOGP8-dependent vulnerabilities
The human-specific nature of NANOGP8 offers a unique advantage for therapeutic development, as targeting this gene would potentially affect cancer cells without disrupting normal stem cell function mediated by the parent NANOG gene. Additionally, approaches targeting NANOGP8 would be expected to have minimal off-target effects on other tissues given its restricted expression pattern .
Preliminary research demonstrating that NANOGP8 promotes cell proliferation suggests that its inhibition could have significant anti-cancer effects, though comprehensive validation studies in multiple cancer models would be required before clinical translation .
Comparative studies between humans and non-human primates offer unique insights into NANOGP8 biology that cannot be gained through other approaches. These studies leverage the human-specific nature of NANOGP8 to understand its contribution to human-specific disease patterns.
The most valuable comparative approaches include:
Cancer susceptibility comparison:
Functional genomics in cross-species contexts:
Introduction of NANOGP8 into non-human primate cells to assess oncogenic potential
Comparison of effects on cell proliferation, stemness, and other cancer-related phenotypes
Assessment of whether NANOGP8 expression alters response to carcinogens or oncogenic drivers
Regulatory network evolution:
Comparison of transcriptional networks between human and non-human primate stem and cancer cells
Investigation of how NANOGP8 insertion may have rewired human-specific regulatory networks
Analysis of whether NANOGP8 complements or competes with NANOG function
Evolutionary genetic studies:
Cross-species tissue culture systems:
Development of matched human and non-human primate cell models
Comparison of cellular response to oncogenic stressors
Assessment of whether NANOGP8 alters cellular plasticity or differentiation potential
These comparative approaches could help address fundamental questions about whether NANOGP8 contributes to human-specific disease susceptibility patterns, potentially explaining part of the higher cancer predisposition in humans compared to other primates . Understanding the specific mechanisms would not only advance cancer biology but could also inform evolutionary medicine approaches to cancer prevention and treatment.
Despite progress in understanding NANOGP8, several critical knowledge gaps remain that limit our comprehensive understanding of its biology and therapeutic potential. Addressing these gaps should be prioritized in future research efforts.
The most significant knowledge gaps include:
Comprehensive expression profiling:
Systematic analysis of NANOGP8 expression across cancer types and subtypes
Single-cell analysis to identify specific cell populations expressing NANOGP8
Understanding of the relationship between NANOGP8 expression and clinical outcomes
Correlation with cancer stem cell markers and stemness properties
Regulatory mechanisms:
Identification of transcription factors and epigenetic regulators controlling NANOGP8 expression
Understanding how the SVA LTR and other NANOGP8-specific regulatory elements function
Elucidation of post-transcriptional regulation mechanisms
Investigation of potential feedback loops between NANOGP8 and other cancer pathways
Functional distinctions from NANOG:
Systematic comparison of NANOGP8 and NANOG target genes and binding sites
Assessment of whether NANOGP8 has gained or lost functions compared to NANOG
Understanding of whether NANOGP8 and NANOG compete, cooperate, or function independently
Structural studies comparing the proteins and their interaction partners
Clinical significance:
Prospective studies correlating NANOGP8 expression with patient outcomes
Investigation of whether NANOGP8 polymorphisms affect cancer risk or treatment response
Assessment of NANOGP8 as a potential biomarker for cancer detection or monitoring
Evaluation of combination therapies targeting NANOGP8-expressing cancers
Evolutionary implications:
Understanding why NANOGP8 became fixed in human populations despite potential cancer risk
Investigation of possible compensatory adaptations that might mitigate NANOGP8-associated cancer risk
Assessment of whether NANOGP8 provides benefits that offset its potential costs
Exploration of NANOGP8's potential role in human-specific traits beyond cancer
Technical challenges:
Addressing these knowledge gaps will require multidisciplinary approaches combining molecular biology, evolutionary genetics, cancer biology, and clinical research. Resolving these uncertainties could significantly advance both our understanding of human-specific cancer biology and our ability to develop targeted therapeutic approaches.
Nanog Homeobox Pseudogene 8 (NANOGP8) is a transcribed retrogene derived from the Nanog homeobox gene. The Nanog gene is crucial for maintaining the pluripotency of embryonic stem cells, and its pseudogenes have been extensively studied for their roles in various biological processes, including cancer cell proliferation .
NANOGP8 is a human-specific retropseudogene that arose through retrotransposition from its parent gene, NANOG. This process involves the reverse transcription of RNA molecules followed by the insertion of DNA copies into the genome. NANOGP8 is unique to humans and is not found in the genomes of other primates, such as chimpanzees .
The evolutionary origin of NANOGP8 is estimated to be between 0.9 to 2.5 million years ago. It arose from a derived variant allele of NANOG containing a 22-nucleotide pair deletion in the 3′ UTR. This variant has remained polymorphic in modern humans .
NANOGP8 is predominantly expressed in cancer cells, where its protein product is tumorigenic. In vitro studies using a recombinant protein have shown that NANOGP8 localizes to the nucleus and can promote cell proliferation, similar to the Nanog protein . This suggests that NANOGP8 may play a role in the reprogramming of cancer cells and contribute to the higher genetic predisposition for cancer in humans compared to other primates .