Next Article in Journal
Impact of Indocyanine Green Dose on Sentinel Lymph Node Mapping in Cervical Cancer: A Systematic Review
Previous Article in Journal
Brain Metastases as Inaugural Sign of Non-Small Cell Lung Carcinoma: Case Series and Review of Literature
Previous Article in Special Issue
Macromutations Yielding Karyotype Alterations (and the Process(es) behind Them) Are the Favored Route of Carcinogenesis and Speciation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Amitotic Cell Division, Malignancy, and Resistance to Anticancer Agents: A Tribute to Drs. Walen and Rajaraman

Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(17), 3106; https://doi.org/10.3390/cancers16173106
Submission received: 16 August 2024 / Revised: 6 September 2024 / Accepted: 6 September 2024 / Published: 8 September 2024
(This article belongs to the Special Issue The Role of Chromosomal Instability in Cancer)

Abstract

:

Simple Summary

Human cells are typically diploid, that is, they contain two sets of chromosomes. Some cells, however, contain three or more sets of chromosomes and are called polyploid. In the early 2000s, Kirsten Walen and Rengaswami Rajaraman and his associates independently reported that polyploid human cells are capable of undergoing asymmetric division via nuclear budding and bursting (amitosis), giving rise to daughter cells that proliferate rapidly and can contribute to tumorigenesis (transformation of normal cells to a neoplastic state). This commentary provides an update on these intriguing discoveries.

Abstract

Cell division is crucial for the survival of living organisms. Human cells undergo three types of cell division: mitosis, meiosis, and amitosis. The former two types occur in somatic cells and germ cells, respectively. Amitosis involves nuclear budding and occurs in cells that exhibit abnormal nuclear morphology (e.g., polyploidy) with increased cell size. In the early 2000s, Kirsten Walen and Rengaswami Rajaraman and his associates independently reported that polyploid human cells are capable of producing progeny via amitotic cell division, and that a subset of emerging daughter cells proliferate rapidly, exhibit stem cell-like properties, and can contribute to tumorigenesis. Polyploid cells that arise in solid tumors/tumor-derived cell lines are referred to as polyploid giant cancer cells (PGCCs) and are known to contribute to therapy resistance and disease recurrence following anticancer treatment. This commentary provides an update on some of these intriguing discoveries as a tribute to Drs. Walen and Rajaraman.

1. Introduction

1.1. Amitotic Cell Division in Polyploid Mammalian Cells

In 2020, the science writer Kerry Benson published a news blog on cancer cell polyploidy highlighting the observations reported by a team at Brown University [1]. Kerry stated that “as researchers and medical professionals work to develop new treatments for cancer, they face a variety of challenges. One is intratumor heterogeneity—the presence of multiple kinds of cancer cells within the same tumor. Often, these “mosaic” tumors include cells, such as polyploidal giant cancer cells, that have evolved to become aggressive and resistant to chemotherapy and radiation.”
In the past, polyploid giant cancer cells (PGCCs) have been largely ignored, said Kerry [1], because PGCCs fail to undergo mitosis, a highly regulated process by which the nuclear and cytoplasmic components of a mother cell are divided into two daughter cells. However, recent studies reported by Xuan et al. [2] have demonstrated that such giant cells can produce rapidly proliferating progeny via amitotic cell division (nuclear budding and bursting similar to simple organisms like fungi). “These cells (emerging from PGCCs) appear to play an active role in invasion and metastasis, so targeting their migratory persistence could limit their effects on cancer progression” noted Michelle Dawson, the senior author of the study [2].
The observation that polyploid human cells can undergo amitotic cell division is not new. In the early 2000s, for example, seminal reports published by Kirsten Walen [3] and Rengaswami Rajaraman and associates (Sundaram et al. [4]) underscored the significance of amitotic cell division of polyploid human cells in the context of tumorigenesis and resistance to anticancer agents.

1.2. Objectives

The purpose of this commentary is to pay tribute to Drs. Walen [3,5,6,7,8,9,10,11] and Rajaraman [4,12,13,14] for their contributions to our understanding of cancer biology in general, and cancer cell repopulation following radio/chemotherapy in particular.
Specifically, this article is arranged as follows: (i) role of polyploidy-amitosis in tumorigenesis (Section 2); (ii) fate of cancer cells with genome instability (Section 3, Section 4, Section 5 and Section 6); and (iii) genome instability in human hereditary cancer-prone disorders, with a focus on Li-Fraumeni syndrome (Section 7).

2. Polyploidy, Amitosis, and In Vitro Cell Transformation

Huang and Zhou have recently published a comprehensive review entitled “DNA damage repair: historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy” [15]. They chronicled the history of the DNA damage and repair journey from the landmark discovery of gene mutation in 1927, the importance of DNA repair pathways in preventing gene mutations and cancer in the 1960s, defining apoptosis in 1972, and the identification of oncogenes, cell cycle checkpoints, and the DNA damage response since the 1980s (also see Figure 1). These discoveries led to the somatic mutation theory of cancer and an approach to cancer therapy purely based on the reductionist theory.
Reductionism is centered on the idea that complex diseases such as cancer can be better explained by breaking them down (“reducing”) into small, simple pieces and studying each piece separately. Accordingly, many groups, our own included (e.g., [16,17,18,19,20]), devoted considerable efforts to characterize signaling pathways that are altered in cancer, to identify the many (hundreds and thousands) of factors that play roles in each pathway (e.g., in the p53 “fireworks” [21,22]), and to conceptualize how these various pathways interact to influence cell fate. The danger of relying on such information-generating approaches to cancer/cancer therapy (high content screens, omics, etc.) has become apparent in the past decade, as discussed by eminent scientists such as Robert Weinberg in 2014 (“Coming Full Circle—From Endless Complexity to Simplicity and Back Again” [23]), Nobel Prize Laureate William Kailen in 2017 (“Publish Houses of Brick, not Mansions of Straw” [24]), Henry Heng in 2019 (“Genome Chaos: Rethinking Genetics, Evolution, and Molecular Medicine” [24], and others (reviewed in [25,26]).
Some observations reported since the 1990s did not support the somatic mutation theory of cancer (e.g., [27] and references therein), which raised the question as to whether mutations beget cancers or cancers beget mutations [27]. To this end, in the early-/mid-2000s Kirsten Walen reported a series of studies demonstrating a non-mutational basis for the transformation of some human cells grown in culture [3,5,6,7]. The experiments were performed with various cell types, including human diploid epithelial cells (i.e., amniocytes) [3,5] and dermal fibroblasts [6,7] that were passaged until they entered the replicative senescence state. Transformation occurred in these cultures either spontaneously or after infection with SV40. The following sequential events in the process of transformation were reported: (i) a small proportion (1–3%) of cells within senescent cultures increased their nuclear contents through endoreduplication; (ii) amitosis/fragmentation of the resulting polyploid nuclei gave rise to multinucleated giant cells; (iii) individual nuclei became surrounded by a cell membrane and exited the multinucleated mother cells via budding; (iv) these daughter cells exhibited extended life span in culture (immortalization) and indicators of neoplastic transformation based on cytogenetic analysis. (The proportion of daughter cells, emerging from each giant cell, that showed these properties was not determined).
Walen’s work continued to shed light on the complexity of tumor initiation and progression that extends far beyond the canonical somatic mutation theory of cancer. In her 2014 article, for example, Walen highlighted the occurrence of parental genome segregation or “gonomery” (a well-known 1/2-size cell volume reduction process in plants) in senescent human polyploid fibroblasts [8]. Gonomery-based functional division in fibroblasts generated near-diploid daughter cells, which showed of a proliferative advantage over the cells of origin. The article was entitled “Neoplastic-Like Cell Changes of Normal Fibroblast Cells Associated with Evolutionary Conserved Maternal and Paternal Genomic Autonomous Behavior…” [8].
As a continuation of this work, Salmina et al. [28] determined how tumor cells segregate their replicated parental genomes which reunite to undergo a meiosis-like recombination after the next replication cycle. (This study was led by Jekaterina Erenpreisa, a long-time colleague of Dr. Walen.) The authors reported that “pseudo-mitosis commonly occurs with a mitotically disabled spindle and closed telomeres, thereby evading practically all of the checkpoints of the mitotic cell cycle. Parental genome segregations with spindle uncoupling has been suggested by Walen in normal human senescing fibroblast cultures and in those treated with spindle poisons, and extrapolated to stem cell biology, highlighting gonomery…as an operating unit of genome segregations” and a mechanism of malignant transformation [28].
In her most recent articles [10,11], Walen discussed her discoveries in relation to epigenetic reprogramming, atavistic mechanisms, and “The First Cell” model of cancer [29]. In her 2021 review [10], Walen stated that “Three-four years ago scientists on the television screen promised cancer-eradication in a 5–10 year time-frame. But now 5 years have gone and the promise of eradication is no longer heard. Why?”
This is an important question. What are the reasons for continuing failures in cancer therapy despite many promises of modern therapeutic strategies (e.g., targeted cancer therapy, precision oncology, synthetic “lethality”) for decades? Perhaps “The assumption of normal mitosis in cancer proliferation is wrong!” said Walen [11]. Other reasons why “The ‘War on Cancer’ Isn’t Yet Won” [30] have been discussed [22,26,31,32,33].

3. Fate of Cancer Cells with Genome Instability

3.1. Amitosis in Solid Tumors/Tumor-Derived Cell Lines

Single cell biology has revealed that solid tumors and tumor-derived cell lines contain a subset of cancer cells with extensive genome instability that are readily distinguishable from the bulk of cancer cells by virtue of their enormous size (reviewed in, e.g., [34,35,36,37,38,39,40,41]). A variety of terms have been used to describe such large-sized cells, including PGCCs, polyaneuploid cancer cells, osteoclast-like cancer cells, pleomorphic cancer cells, blastomere cancer cells, and multinucleated cancer cells [41]. For simplicity, we will refer to giant cells with a highly enlarged nucleus, multiple nuclei, and/or multiple micronuclei as PGCCs.
Key discoveries reported between 1956 and the mid-2000s, which have been particularly useful for our group to understand the significance of PGCCs in tumorigenesis and therapy resistance of solid tumors (see, e.g., [42,43,44]), are pointed out below. Figure 2 is reproduced from our 2008 review [42].
In 1956, Puck and Marcus demonstrated that exposure of HeLa (cervical carcinoma) cells to ionizing radiation results in the creation of multinucleated giant cells that fail to form macroscopic colonies (aggregates of at least 50 cells) within ~10 days post-irradiation [45] (also see Appendix A). These giant cells were shown to remain adherent to the culture dish for long times (weeks) after irradiation and exhibited the ability to secrete growth-promoting factors.
The significance of the seminal observations reported by Puck and Marcus regarding giant cells was largely overlooked by many research groups, including our own [19], perhaps because such cells cease to proliferate or proliferate at a very slow rate, and thus are often misrepresented as “dead” in the colony formation and other widely used radiosensitivity and chemosensitivity assays [46,47].
In 2000, however, Erenpreisa et al. [48] and Illedge et al. [49] reported that giant cells that emerge in cultures of Burkitt’s lymphoma cells, in response to ionizing radiation exposure (10 Gy), can undergo depolyploidization and give rise to progeny that enter the mitotic cycle. This depolyploidization process was subsequently shown by these authors to involve key mediators of meiosis, self-renewal, and mitosis (reviewed in [34,50]).
As mentioned above, in 2002, Kirsten Walen [3] demonstrated that giant human cells can also give rise to progeny via nuclear budding and bursting (amitosis). In 2004, Rengaswami Rajaraman and his associates [4] reported a similar observation based on live-imaging studies and coined the term “neosis” for this parasexual somatic reduction process of cell division. Rapidly proliferating cells that emerged from giant cells were called “karyoplasts” [3] or “Raju” cells [4] and were shown to exhibit stem cell-like properties.
In 2004, Navolanic and associates discussed the potential implications of these discoveries in a commentary entitled “Neosis and its Potential Role in Cancer Development and Chemoresistance” [51]. These authors stated that “cell division in eukaryotes by any other process than mitosis or meiosis is certainly a radical concept. Justifiably, the prospect of neosis will be regarded with skepticism unless the claims of the authors can be verified in other laboratories” [51]. In the past decade, numerous groups have indeed verified neosis in different human cell types (e.g., [2,52,53,54,55,56,57,58,59,60,61,62,63]).

3.2. Amitosis and System-Level Information Alteration

In the past decade, several reviews have discussed the relationship between genome chaos, new system creation, and evolution [33,64,65,66,67,68]. These include an article entitled “The New Era of Cancer Cytogenetics and Cytogenomics” that highlights the importance of profiling the karyotype (rather than solely profiling gene mutations) especially in cancer, where karyotype alterations contribute to cellular macroevolution [67]. The link between amitotic cell division and the concept of karyotype coding and other means of system-level information alteration remains to be determined.

4. Targeting PGCCs in Cancer Therapy

The mechanisms of the formation and fate of PGCCs as well as their prevalence and prognostic value in different types of solid tumors are now well established and extensively discussed [34,35,36,37,38,39,40,41]. Some recent discoveries that suggest the potential of targeting PGCCs for improving patient outcomes are outlined below.
  • The RhoA-Rock1 pathway, vimentin filaments, and overall actin cytoskeletal network drive the increased stiffness and migratory persistence of PGCCs [2,61,69].
  • The formation of PGCCs and their tumor repopulating progeny (via neosis) can be blocked by the contraceptive drug mifepristone [70].
  • PGCCs developed following cisplatin treatment have a high content of mitochondria and a distinct metabolic profile, which includes high levels of lipid droplets and cholesterol. These PGCCs could be targeted using zoledronic acid, a potent inhibitor of osteoclasts (multinucleated bone cells) [71], which is commonly used to treat osteoporosis and bone metastases [72,73].
  • Treatment with LCL521 or simvastatin disrupts cholesterol signaling and interferes with PGCC progeny formation [74].
  • The sphingolipid enzyme acid ceramidase (ASAH1) is required for the generation of progeny from PGCCs [54,56], and this process (neosis) can be inhibited by tamoxifen, which exerts an off-target effect on ASAH1 [56].
  • Treatment with UC2288 reduces acid ceramide expression and inhibits depolyploidization of PGCCs [75]. UC2288 is an attenuator of p21WAF1 (p21) [76], which also exerts off-target effects on the EGFR/ERK (epidermal growth factor receptor/extracellular signal-regulated kinase) signaling pathway [77].
  • The ESCRT (endosomal complexes required for transport) proteins are involved in the budding of PGCCs [78]. Treatment of PGCCs with interferon, a modulator of ESCRT, prevented PGCC budding [78]. In that study, PGCCs were created following exposure to ionizing radiation and were referred to as “radiation-tolerant persister” cells.
  • The expression of PLN4, a perilipin involved in lipid droplet stability, contributes to resistance to cytotoxic therapy in some patients with triple negative breast cancer [79]. PLN4-expressing PGCCs are enriched in tumors of these patients [79].
Some of these discoveries and their clinical implications were discussed in recent blogs entitled “Study discovers potential target for treating aggressive cancer cells” by Kerry Benson [1], “Targeting monster cancer cells could reduce recurrence rates after cancer therapy” by Emma Funk [80], and “First study on physical properties of giant cancer cells may inform new treatments…” by Mollie Rappe [81].
Disappointingly, most of these recent reports (including blogs) did not acknowledge the fact that Walen, Rajaraman and others demonstrated the significance of polyploidy-amitosis-neosis in tumorigenesis/therapy resistance about two decades ago.
Time will tell whether these discoveries will lead to therapeutic strategies that can be adapted in the clinic to improve the outcome of treating patients with solid tumors. To this end, it is important to note that PGCCs represent only one piece of the complex puzzle of therapy resistance and relapse [22,46].

5. The Role of PGCCs in Minimal Residual Disease and Tumor Repopulation Post-Therapy

Preclinical studies have identified four phases of solid tumor therapy following exposure to ionizing radiation (e.g., [78]) and chemotherapeutic drugs (e.g., cisplatin [82,83]) that are consistent with common trends in patient outcomes [84]. These phases are outlined below. Figure 3 is adapted from a comprehensive review on polyploidy/neosis by Zhao et al. [85] and a recent report by that group [78] in which they determined the basis for the minimal residual disease (MRD) in the course of radiotherapy. The cartoons showing cell images are redrawn from the time-lapse microscopy data presented in the latter article (see Figure 3B in [78]).
i.
Treatment Phase. This first phase involves conventional therapeutic strategies (such as surgery, chemotherapy, and radiation therapy) to inhibit tumor growth and hopefully to prevent or at least mitigate metastasis. The majority (perhaps >95%) of anticancer studies have focused on this vital phase of cancer therapy.
ii.
Response. A proportion of cancer cells within a solid tumor responds to genotoxic insult (incurred from the treatment phase) by entering a state of dormancy (active sleep). This group includes PGCCs. In tissue culture studies, it may take ~10 days after treatment for PGCCs to be fully manifested (see, e.g., [78]). It is worth noting that ubiquitously used preclinical anticancer end-points (e.g., multiwell plate cell “viability” and tumor growth delay assays in live animals) are performed within this time frame, and thus they often overlook the impact of PGCCs or score them as “dead” (see, e.g., [22,26]).
iii.
Dormancy. Senescence-like dormancy [84] (also referred to as MRD [85]) is an extended latency period during which PGCCs undergo depolyploidization through a meiosis-like process as well as through amitosis-neosis. PGCCs are also known to have the ability to transfer a small portion of their nuclear material containing stem cell markers to neighboring cells via cytoplasmic tunnels [86].
iv.
Recurrence. Rapidly proliferating progenitor cells emerging from PGCCs repopulate the tumor. Such cells have stem cell characteristics and can show resistance to conventional therapies (used in the “treatment phase”). Whether cells subjected to horizontal gene transfer also exhibit these properties is not known.
The take home message is loud and clear: the initial solid tumor and recurrent tumor post-therapy are different “diseases” requiring different treatment strategies.

6. Amitotic Cell Division Contributes to Intratumor Heterogeneity

Intratumor heterogeneity refers to the presence of different subpopulations of cancer cells within a solid tumor/tumor-derived cell line that exhibit therapy resistance via various molecular and cellular processes. In the 2000s, the presence of cancer stem cells within a solid tumor was emerging as a key contributor to this heterogeneity. As we pointed out in the late 2000s [42,43,44], “another prominent source of cancer cell heterogeneity is via karyoplasts emerging from polyploid/multinucleated giant cells through neotic cell division...Like cancer stem cells, cancer karyoplasts exhibit a high degree of resistance to the cytotoxic effects of cancer therapeutic agents” [42]. Since then, a number of other genetic and non-genetic processes have been demonstrated to contribute to this heterogeneity. These include: (i) reversible dormancy via premature senescence [87,88,89,90]; (ii) reversible dormancy reflecting drug-tolerant persister cells, both preexisting and therapy induced [91,92]; (iii) cancer cell survival subsequent to engagement of apoptosis and other regulated cell death pathways [22,91,93]; and (iv) heterogeneity of p53 protein expression, which “may represent p53 mutant patches indicative of clonal expansion, epigenetic modifications, and/or a number of other possibilities” [94].

7. Human Genetic Disorders Associated with Genome Instability and Cancer Predisposition: Does Amitosis-Neosis Play a Role?

In the aforementioned review on the “DNA damage repair journey” by Huang and Zhou [15], the authors provided a brief history of xeroderma pigmentosum, an autosomal recessive cancer-prone disorder characterized by DNA repair deficiency. Table 1 contains some clinical and molecular features of this and other extensively studied human hereditary disorders, all of which are characterized by genome instability (for details, see [43]). The relationship (if any) between amitosis/neosis and some clinical features (e.g., cancer proneness) of these disorders remains unknown.
Our group has contributed to the understanding of the DNA damage response of some of these cancer-prone disorders (reviewed in [95]). Of particular relevance to the current discussion, we have observed a high frequency of polyploid/multinucleated giant cells in dermal fibroblast strains from affected members of a cancer-prone family with the Li-Fraumeni syndrome (LFS) [96]. Giant cells were enriched in LFS fibroblasts as a function of in vitro culture age, peaking at passage 38 (Figure 4), or in young (low passage) cultures after exposure to ionizing radiation. We also provided evidence suggesting that the p16INK4A tumor suppressor might play a key role in preventing the genesis of metastatic progeny from polyploid/multinucleated giant cells (i.e., by preventing neosis) [96].
Unfortunately, we were unable to secure funds to explore this intriguing hypothesis because, in those days (prior to 2014 [31]), PGCCs were generally considered to reflect artifacts that may only occur in cell lines maintained in culture. In addition, as recently pointed out by Pienta et al. [36], “the majority of cancer research and treatment development communities have disregarded these (giant) cells as irreversibly senescent or destined for mitotic catastrophe and death.”
The significant roles played by giant cells in tumorigenesis and therapy resistance are becoming increasingly appreciated, which serves to underscore the pioneering contributions of Walen and Rajaraman to the polyploidy-amitosis-neosis field about two decades ago.

8. Conclusions

Studies reported since the 1950s have revealed that moderate, clinically relevant doses of ionizing radiation and chemotherapeutic drugs trigger the generation of cells with various manifestations of genome chaos, including PGCCs, that remain viable, secrete growth-promoting factors, and are capable of producing progeny with stem cell-like properties. PGCCs can contribute to tumor repopulation by various means, including: (i) depolyploidization, through which PGCCs reduce their genome to a size (paradiploid) that is compatible with mitosis; (ii) amitotic cell division, which results in the emergence of numerous (hundreds to thousands) of proliferating daughter cells from a single PGCC; and (iii) horizontal gene transfer via cytoplasmic tunnels by which PGCCs can export a small portion of their nuclear material containing stem cell markers to neighboring cells (reviewed in, e.g., [26,46]).
Surprisingly, PGCCs continue to be overlooked in most cancer-related articles that discuss the “bright” (pro-death) side of apoptosis in solid tumor therapy [97,98,99,100,101], the “dark” (pro-survival) side of apoptosis in cancer therapy [91,93], targeting the DNA damage response (e.g., the p53/p21 pathway) in cancer therapy [102,103,104,105,106,107], as well as numerous articles that highlight limitations of implementing anticancer strategies under the term “precision oncology” (reviewed in [22,26]).
When discussing the possible reasons for this serious oversight (disregarding PGCCs) with the senior author of a comprehensive review on breast cancer, he replied in an email communication that “we are all learning… According to the 19th century German philosopher Arthur Schopenhauer, All truth passes through three stages: First, it is ridiculed. Second, it is violently opposed. Third, it is accepted as self-evident”.
The lessons that we have learned from landmark discoveries of Walen, Rajaraman, Erenpreisa, and other pioneering PGCC biologists are invaluable, and will hopefully lead to improved efficacy of conventional solid tumor therapies.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflicts of interest.

Appendix A

Figure A1. The development and fate of multinucleated giant cells following exposure to ionizing radiation, as reported by Puck and Marcus over 60 years ago [45]. (For details, see [46]).
Figure A1. The development and fate of multinucleated giant cells following exposure to ionizing radiation, as reported by Puck and Marcus over 60 years ago [45]. (For details, see [46]).
Cancers 16 03106 g0a1

References

  1. Benson, K. Study Discovers Potential Target for Treating Aggressive Cancer Cells. 2021. Available online: https://www.ikcest.org/articleS-552496.htm (accessed on 10 August 2024).
  2. Xuan, B.; Ghosh, D.; Jiang, J.; Shao, R.; Dawson, M.R. Vimentin filaments drive migratory persistence in polyploidal cancer cells. Proc. Natl. Acad. Sci. USA 2020, 117, 26756–26765. [Google Scholar] [CrossRef] [PubMed]
  3. Walen, K.H. The origin of transformed cells: Studies of spontaneous and induced cell transformation in cell cultures from marsupials, a snail, and human amniocytes. Cancer Genet. 2002, 133, 45–54. [Google Scholar] [CrossRef]
  4. Sundaram, M.; Guernsey, D.L.; Rajaraman, M.M.; Rajaraman, R. Neosis: A novel type of cell division in cancer. Cancer Biol. Ther. 2004, 3, 207–218. [Google Scholar] [CrossRef] [PubMed]
  5. Walen, K.H. Spontaneous cell transformation: Karyoplasts derived from multinucleated cells produce new cell growth in senescent human epithelial cell cultures. Cell. Dev. Biol. Anim. 2004, 40, 150–158. [Google Scholar] [CrossRef]
  6. Walen, K.H. Budded karyoplasts from multinucleated fibroblast cells contain centrosomes and change their morphology to mitotic cells. Cell Biol. Int. 2005, 29, 1057–1065. [Google Scholar] [CrossRef]
  7. Walen, K.H. Genetic stability of senescence reverted cells: Genome reduction division of polyploidy cells, aneuploidy and neoplasia. Cell Cycle 2008, 7, 1623–1629. [Google Scholar] [CrossRef]
  8. Walen, K.H. Neoplastic-like cell changes of normal fibroblast cells associated with evolutionary conserved maternal and paternal genomic autonomous behavior (gonomery). J. Cancer Ther. 2014, 05, 860–877. [Google Scholar] [CrossRef]
  9. Walen, K.H. Genomic instability in cancer II: 4N-skewed (90°) reductive division via fragile sites to fitness increase for solid and hematological cancer beginnings. J. Cancer Ther. 2019, 10, 537–564. [Google Scholar] [CrossRef]
  10. Walen, K.H. A traceable cancer model: DNA damage, fragile site-SMGs, mitotic slippage, 4n-genome-reduction to fitness-gained, initiating, 2n first cells. J. Cancer Ther. 2021, 12, 365–386. [Google Scholar] [CrossRef]
  11. Walen, K.H. Epigenetic enabled normal human cells, lead to first cell’s unique division system, driving tumorigenesis evolution. J. Cancer Ther. 2022, 13, 48–69. [Google Scholar] [CrossRef]
  12. Rajaraman, R.; Rajaraman, M.M.; Rajaraman, S.R.; Guernsey, R.L. Neosis—A paradigm of self-renewal in cancer. Cell Biol. Int. 2005, 29, 1084–1097. [Google Scholar] [CrossRef] [PubMed]
  13. Rajaraman, R.; Guernsey, D.L.; Rajaraman, M.M.; Rajaraman, S.R. Stem cells, senescence, neosis and self-renewal in cancer. Cell Biol. Int. 2006, 29, 1084–1097. [Google Scholar] [CrossRef] [PubMed]
  14. Rajaraman, R.; Guernsey, D.L.; Rajaraman, M.M.; Rajaraman, S.R. Neosis—A parasexual somatic reduction division in cancer. Int. J. Hum. Genet. 2007, 7, 29–48. [Google Scholar] [CrossRef]
  15. Huang, R.; Zhou, P.K. DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 254. [Google Scholar] [CrossRef]
  16. Mirzayans, R.; Waters, R. DNA damage and its repair in human normal or xeroderma pigmentosum fibroblasts treated with 4-nitroquinoline 1-oxide or its 3-methyl derivative. Carcinogenesis 1981, 2, 1359–1362. [Google Scholar] [CrossRef]
  17. Mirzayans, R.; Famulski, K.S.; Enns, L.; Fraser, M.; Paterson, M.C. Characterization of the signal transduction pathway mediating γ ray-induced inhibition of DNA synthesis in human cells: Indirect evidence for involvement of calmodulin but not protein kinase C nor p53. Oncogene 1995, 11, 1597–1605. [Google Scholar]
  18. Mirzayans, R.; Enns, L.; Dietrich, K.; Barley, R.D.; Paterson, M.C. Faulty DNA polymerase δ/ɛ-mediated excision repair in response to γ radiation or ultraviolet light in p53-deficient fibroblast strains from affected members of a cancer-prone family with Li-Fraumeni syndrome. Carcinogenesis 1996, 17, 691–698. [Google Scholar] [CrossRef]
  19. Barley, R.D.C.; Enns, L.; Paterson, M.C.; Mirzayans, R. Aberrant p21WAF1-dependent growth arrest as the possible mechanism of abnormal resistance to ultraviolet light cytotoxicity in Li-Fraumeni syndrome fifibroblast strains heterozygous for TP53 mutations. Oncogene 1998, 17, 533–543. [Google Scholar] [CrossRef]
  20. Murray, D.; Mirzayans, R.; Scott, A.L.; Allalunis-Turner, M.J. Influence of oxygen on the radiosensitivity of human glioma cell lines. Am. J. Clin. Oncol. 2003, 26, e169–e177. [Google Scholar] [CrossRef]
  21. Uversky, V.N. p53 proteoforms and intrinsic disorder: An illustration of the protein structure-function continuum concept. Int. J. Mol. Sci. 2016, 17, 1874. [Google Scholar] [CrossRef]
  22. Mirzayans, R. Changing the landscape of solid tumor therapy from apoptosis-promoting to apoptosis-inhibiting strategies. Curr. Issues Mol. Biol. 2024, 46, 5379–5396. [Google Scholar] [CrossRef]
  23. Weinberg, R.A. Coming full circle-from endless complexity to simplicity and back again. Cell 2014, 157, 267–273. [Google Scholar] [CrossRef] [PubMed]
  24. Kailen, W.G. Publish houses of brick, not mansions of straw. Nature 2017, 5454, 387. [Google Scholar]
  25. Heng, H.H. Genome Chaos: Rethinking Genetics, Evolution, and Molecular Medicine; Academic Press: Cambridge, MA, USA, 2019; p. 556. [Google Scholar]
  26. Mirzayans, R.; Murray, D. What are the reasons for continuing failures in cancer therapy? Are misleading/inappropriate preclinical assays to be blamed? Might some modern therapies cause more harm than benefit? Int. J. Mol. Sci. 2022, 23, 13217. [Google Scholar] [CrossRef]
  27. Prehn, R.T. Cancers beget mutations versus mutations beget cancers. Cancer Res. 1994, 54, 5296–5300. [Google Scholar] [PubMed]
  28. Salmina, K.; Huna, A.; Kalejs, M.; Pjanova, D.; Scherthan, H.; Cragg, M.S.; Erenpreisa, J. The cancer aneuploidy paradox: In the light of evolution. Genes 2019, 10, 83. [Google Scholar] [CrossRef]
  29. Raza, A. The First Cell: And the Human Costs of Pursuing Cancer to the Last; Basic Books: New York, NY, USA, 2019; p. 368. [Google Scholar]
  30. Editorial. The ‘war on cancer’ isn’t yet won. Nature 2022, 601, 297. [Google Scholar] [CrossRef]
  31. Coward, J.; Harding, A. Size does matter: Why polyploid tumor cells are critical drug targets in the war on cancer. Front. Oncol. 2014, 4, 123. [Google Scholar] [CrossRef]
  32. Maeda, H.; Khatami, M. Analyses of repeated failures in cancer therapy for solid tumors: Poor tumor-selective drug delivery, low therapeutic efficacy and unsustainable costs. Clin. Transl. Med. 2018, 7, 11. [Google Scholar] [CrossRef]
  33. Heng, J.; Heng, H.H. Genome chaos, information creation, and cancer emergence: Searching for new frameworks on the 50th anniversary of the “war on cancer”. Genes 2022, 13, 101. [Google Scholar] [CrossRef]
  34. Erenpreisa, J.; Salmina, K.; Huna, A.; Jackson, T.R.; Vazquez-Martin, A.; Cragg, M.S. The “virgin birth”, polyploidy, and the origin of cancer. Oncoscience 2015, 2, 3–14. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, J.; Niu, N.; Zhang, J.; Qi, L.; Shen, W.; Donkena, K.V.; Feng, Z.; Liu, J. Polyploid giant cancer cells (PGCCs): The evil roots of cancer. Curr. Cancer Drug Targets 2019, 19, 360–367. [Google Scholar] [CrossRef]
  36. Pienta, K.J.; Hammarlund, E.U.; Austin, R.H.; Axelrod, R.; Brown, J.S.; Amend, S.R. Cancer cells employ an evolutionarily conserved polyploidization program to resist therapy. Semin. Cancer Biol. 2022, 81, 145–159. [Google Scholar] [CrossRef] [PubMed]
  37. Saini, G.; Joshi, S.; Garlapati, C.; Li, H.; Kong, J.; Krishnamurthy, J.; Reid, M.D.; Aneja, R. Polyploid giant cancer cell characterization: New frontiers in predicting response to chemotherapy in breast cancer. Semin. Cancer Biol. 2022, 81, 220–231. [Google Scholar] [CrossRef]
  38. Liu, J.; Niu, N.; Li, X.; Zhang, X.; Sood, A.K. The life cycle of polyploid giant cancer cells and dormancy in cancer: Opportunities for novel therapeutic interventions. Semin. Cancer Biol. 2022, 81, 132–144. [Google Scholar] [CrossRef]
  39. Sikora, E.; Czarnecka-Herok, J.; Bojko, A.; Sunderland, P. Therapy-induced polyploidization and senescence: Coincidence or interconnection? Semin. Cancer Biol. 2022, 81, 83–95. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, J.; Qiao, Q.; Xu, H.; Zhou, R.; Liu, X. Human cell polyploidization: The good and the evil. Semin. Cancer Biol. 2022, 81, 54–63. [Google Scholar] [CrossRef]
  41. Zhou, X.; Zhou, M.; Zheng, M.; Tian, S.; Yang, X.; Ning, Y.; Li, Y.; Zhang, S. Polyploid giant cancer cells and cancer progression. Front. Cell Dev. Biol. 2022, 10, 1017588. [Google Scholar] [CrossRef]
  42. Mirzayans, R.; Murray, D. Pharmacological modulation of p53 function in cancer therapy. Curr. Signal. Transduct. Ther. 2008, 3, 183–194. [Google Scholar] [CrossRef]
  43. Mirzayans, R.; Murray, D. Human genetic disorders associated with genome instability, premature aging and cancer predisposition. Open Cancer J. 2008, 2, 42–52. [Google Scholar] [CrossRef]
  44. Mirzayans, R.; Murray, D. Cellular Senescence: Implications for Cancer Therapy; Monograph Garvey, R.B., Ed.; Nova Science Publishers, Inc.: Hauppauge, NY, USA, 2009; p. 130. [Google Scholar]
  45. Puck, T.T.; Marcus, P.I. Action of X-rays on mammalian cells. J. Exp. Med. 1956, 103, 653–666. [Google Scholar] [CrossRef] [PubMed]
  46. Mirzayans, R.; Murray, D. Intratumor heterogeneity and treatment resistance of solid tumors with a focus on polyploid/senescent giant cancer cells (PGCCs). Int. J. Mol. Sci. 2023, 24, 11534. [Google Scholar] [CrossRef]
  47. Eastman, A. Improving anticancer drug development begins with cell culture: Misinformation perpetuated by the misuse of cytotoxicity assays. Oncotarget 2017, 8, 8854–8866. [Google Scholar] [CrossRef]
  48. Erenpreisa, J.A.; Cragg, M.S.; Fringes, B.; Sharakhov, I.; Illidge, T.M. Release of mitotic descendants by giant cells from irradiated Burkitt’s lymphoma cell lines. Cell Biol. Int. 2000, 24, 635–648. [Google Scholar] [CrossRef]
  49. Illidge, T.M.; Cragg, M.S.; Fringes, B.; Olive, P.; Erenpresia, J.A. Polyploid giant cells provide a survival mechanism of p53 mutant cells after DNA damage. Cell Biol. Int. 2000, 24, 621–633. [Google Scholar] [CrossRef]
  50. Erenpreisa, J.; Cragg, M.S. MOS, aneuploidy and the ploidy cycle of cancer cells. Oncogene 2010, 29, 5447–5451. [Google Scholar] [CrossRef] [PubMed]
  51. Navolanic, P.M.; Akula, S.M.; McCubrey, J.A. Neosis and its potential role in cancer development and chemoresistance. Cancer Biol. Ther. 2004, 3, 219–220. [Google Scholar] [CrossRef]
  52. Zhang, S.; Mercado-Uribe, I.; Xing, Z.; Sun, B.; Kuang, J.; Liu, J. Generation of cancer stem-like cells through the formation of polyploid giant cancer cells. Oncogene 2014, 33, 116–128. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, D.; Yang, X.; Yang, Z.; Fei, F.; Li, S.; Qu, J.; Zhang, M.; Li, Y.; Zhang, X.; Zhang, S. Daughter cells and erythroid cells budding from PGCCs and their clinicopathological significances in colorectal cancer. J. Cancer 2017, 8, 469–478. [Google Scholar] [CrossRef]
  54. White-Gilbertson, S.; Lu, P.; Norris, J.S.; Voelkel-Johnson, C. Genetic and pharmacological inhibition of acid ceramidase prevents asymmetric cell division by neosis. J. Lipid Res. 2019, 60, 1225–1235. [Google Scholar] [CrossRef]
  55. Fei, F.; Liu, K.; Li, C.; Du, J.; Wei, Z.; Li, B.; Li, Y.; Zhang, Y.; Zhang, S. Molecular mechanisms by which S100A4 regulates the migration and invasion of PGCCs with their daughter cells in human colorectal cancer. Front. Oncol. 2020, 10, 182. [Google Scholar] [CrossRef]
  56. White-Gilbertson, S.; Lu, P.; Jones, C.M.; Chiodini, S.; Hurley, D.; Das, A.; Delaney, J.R.; Norris, J.S.; Voelkel-Johnson, C. Tamoxifen is a candidate first-in-class inhibitor of acid ceramidase that reduces amitotic division in polyploid giant cancer cells—Unrecognized players in tumorigenesis. Cancer Med. 2020, 9, 3142–3152. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, Z.; Feng, X.; Deng, Z.; Cheng, J.; Wang, Y.; Zhao, M.; Zhao, Y.; He, S.; Huang, Q. Irradiation-induced polyploid giant cancer cells are involved in tumor cell repopulation via neosis. Mol. Oncol. 2021, 15, 2219–2234. [Google Scholar] [CrossRef]
  58. Zhao, Q.; Zhang, K.; Li, Z.; Zhang, H.; Fu, F.; Fu, J.; Zheng, M.; Zhang, S. High migration and invasion ability of PGCCs and their daughter cells associated with the nuclear localization of S100A10 modified by SUMOylation. Front. Cell Dev. Biol. 2021, 9, 696871. [Google Scholar] [CrossRef] [PubMed]
  59. Fu, F.; Chen, L.; Yang, X.; Fan, L.; Zhang, M.; Chen, S.; Zheng, M.; Gao, M.; Zhang, S. PLK4 is a key molecule in the formation of PGCCs and promotes invasion and migration of progeny cells derived from PGCCs. J. Cancer 2022, 13, 2954–2969. [Google Scholar] [CrossRef] [PubMed]
  60. Liu, Y.; Shi, Y.; Wu, M.; Liu, J.; Wu, H.; Xu, C.; Chen, L. Hypoxia-induced polypoid giant cancer cells in glioma promote the transformation of tumor-associated macrophages to a tumor-supportive phenotype. CNS Neurosci. Ther. 2022, 28, 1326–1338. [Google Scholar] [CrossRef]
  61. Peña, C.M.; Skipper, T.A.; Hsu, J.; Schechter, I.; Ghosh, D.; Dawson, M.R. Metronomic and single high-dose paclitaxel treatments produce distinct heterogenous chemoresistant cancer cell populations. Sci. Rep. 2023, 13, 19232. [Google Scholar]
  62. Zhou, M.; Ma, Y.; Chiang, C.C.; Rock, E.C.; Butler, S.C.; Anne, R.; Yatsenko, S.; Gong, Y.; Chen, Y.C. Single-cell morphological and transcriptome analysis unveil inhibitors of polyploid giant breast cancer cells in vitro. Commun. Biol. 2023, 6, 1301. [Google Scholar] [CrossRef]
  63. Fan, L.; Zheng, M.; Zhou, X.; Yu, Y.; Ning, Y.; Fu, W.; Xu, J.; Zhang, S. Molecular mechanism of vimentin nuclear localization associated with the migration and invasion of daughter cells derived from polyploid giant cancer cells. J. Transl. Med. 2023, 21, 719. [Google Scholar] [CrossRef]
  64. Ye, C.J.; Sharpe, Z.; Alemara, S.; Mackenzie, S.; Liu, G.; Abdallah, B.; Horne, S.; Regan, S.; Heng, H.H. Micronuclei and genome chaos: Changing the system inheritance. Genes 2019, 10, 366. [Google Scholar] [CrossRef]
  65. Noble, D. Cellular Darwinism: Regulatory networks, stochasticity, and selection in cancer development. Prog. Biophys. Mol. Biol. 2021, 165, 66–71. [Google Scholar] [CrossRef]
  66. Gecow, A.; Iantovics, L.B.; Tez, M. Cancer and chaos and the complex network model of a multicellular organism. Biology 2022, 11, 1317. [Google Scholar] [CrossRef] [PubMed]
  67. Ye, J.C.; Heng, H.H. The new era of cancer cytogenetics and cytogenomics. Methods Mol. Biol. 2024, 2825, 3–37. [Google Scholar] [PubMed]
  68. Kasperski, A.; Heng, H.H. The spiral model of evolution: Stable life forms of organisms and unstable life forms of cancers. Int. J. Mol. Sci. 2024, 25, 9163. [Google Scholar] [CrossRef]
  69. Xuan, B.; Ghosh, D.; Cheney, E.M.; Clifton, E.M.; Dawson, M.R. Dysregulation in actin cytoskeletal organization drives increased stiffness and migratory persistence in polyploidal giant cancer cells. Sci. Rep. 2018, 8, 11935. [Google Scholar] [CrossRef]
  70. Zhang, X.; Yao, Y.; Li, X.; Niu, N.; Liu, Y.; Hajek, R.A.; Peng, G.; Westin, S.; Sood, A.K.; Liu, J. Targeting polyploid giant cancer cells potentiates a therapeutic response and overcomes resistance to PARP inhibitors in ovarian cancer. Sci. Adv. 2023, 9, eadf7195. [Google Scholar] [CrossRef]
  71. Adibi, R.; Moein, S.; Gheisari, Y. Zoledronic acid targets chemo-resistant polyploid giant cancer cells. Sci. Rep. 2023, 13, 419. [Google Scholar] [CrossRef]
  72. Russell, R.G. Bisphosphonates: The first 40 years. Bone 2011, 49, 2–19. [Google Scholar] [CrossRef] [PubMed]
  73. Cremers, S.; Drake, M.T.; Ebetino, F.H.; Bilezikian, J.P.; Russell, R.G.G. Pharmacology of bisphosphonates. Br. J. Clin. Pharmacol. 2019, 85, 1052–1062. [Google Scholar] [CrossRef]
  74. White-Gilbertson, S.; Lu, P.; Esobi, I.; Echesabal-Chen, J.; Mulholland, P.J.; Gooz, M.; Ogretmen, B.; Stamatikos, A.; Voelkel-Johnson, C. Polyploid giant cancer cells are dependent on cholesterol for progeny formation through amitotic division. Sci. Rep. 2022, 12, 8971. [Google Scholar] [CrossRef]
  75. White-Gilbertson, S.; Lu, P.; Saatci, O.; Sahin, Z.; Delaney, J.R.; Ogretmen, B.; Voelkel-Johnson, C. Transcriptome analysis of polyploid giant cancer cells and their progeny reveals a functional role for p21 in polyploidization and depolyploidization. J. Biol. Chem. 2024, 300, 107136. [Google Scholar] [CrossRef]
  76. Wettersten, H.I.; Hwang, S.H.; Li, C.; Shiu, E.Y.; Wecksler, A.T.; Hammock, B.D.; Weiss, R.H. A novel p21 attenuator which is structurally related to sorafenib. Cancer Biol. Ther. 2013, 14, 278–285. [Google Scholar] [CrossRef] [PubMed]
  77. Liang, R.; Zhu, X. UC2288 induces cell apoptosis of nasopharyngeal carcinoma cells via inhibiting EGFR/ERK pathway. Cancer 2021, 12, 988–995. [Google Scholar] [CrossRef] [PubMed]
  78. Zhao, Y.; Lu, T.; Song, Y.; Wen, Y.; Deng, Z.; Fan, J.; Zhao, M.; Zhao, R.; Luo, Y.; Xiem, J.; et al. Cancer cells enter an adaptive persistence to survive radiotherapy and repopulate tumor. Adv. Sci. 2023, 10, 2204177. [Google Scholar] [CrossRef] [PubMed]
  79. Sirois, I.; Aguilar-Mahecham, A.; Lafleur, J.; Fowler, M.; Vu, V.; Scriver, M.; Buchanan, M.; Chabot, C.; Ramanathan, A.; Balachandran, B.; et al. A unique morphological phenotype in chemoresistant triple-negative breast cancer reveals metabolic reprogramming and PLIN4 expression as a molecular vulnerability. Mol. Cancer Res. 2019, 17, 2492–2507. [Google Scholar] [CrossRef]
  80. Funk, E. Targeting Monster Cancer Cells Could Reduce Recurrence Rates after Cancer Therapy. 2024. Available online: https://web.musc.edu/about/news-center/2024/05/28/monster-cancer-cells (accessed on 10 August 2024).
  81. Rappe, M. First Study on Physical Properties of Giant Cancer Cells May Inform New Treatments. 2018. Available online: https://www.brown.edu/news/2018-08-13/giantcells (accessed on 10 August 2024).
  82. Puig, P.E.; Guilly, M.N.; Bouchot, A.; Droin, N.; Cathelin, D.; Bouyer, F.; Favier, L.; Ghiringhelli, F.; Kroemer, G.; Solary, E.; et al. Tumor cells can escape DNA-damaging cisplatin through DNA endoreduplication and reversible polyploidy. Cell Biol. Int. 2008, 32, 1031–1043. [Google Scholar] [CrossRef]
  83. Weng, C.H.; Wu, C.S.; Wu, J.C.; Kung, M.L.; Wu, M.H.; Tai, M.H. Cisplatin-induced giant cells formation is involved in chemoresistance of melanoma cells. Int. J. Mol. Sci. 2020, 21, 7892. [Google Scholar] [CrossRef]
  84. Qin, S.; Jiang, J.; Lu, Y.; Nice, E.C.; Huang, C.; Zhang, J.; He, W. Emerging role of tumor cell plasticity in modifying therapeutic response. Signal Transduct. Target. Ther. 2020, 5, 228. [Google Scholar] [CrossRef]
  85. Zhao, Y.; He, S.; Zhao, M.; Huang, Q. Surviving the storm: The role of poly- and depolyploidization in tissues and tumors. Adv. Sci. 2024, 11, 2306318. [Google Scholar] [CrossRef]
  86. Díaz-Carballo, D.; Saka, S.; Klein, J.; Rennkamp, T.; Acikelli, A.H.; Malak, S.; Jastrow, H.; Wennemuth, G.; Tempfer, C.; Schmitz, I.; et al. A distinct oncogenerative multinucleated cancer cell serves as a source of stemness and tumor heterogeneity. Cancer Res. 2018, 78, 2318–2331. [Google Scholar] [CrossRef]
  87. Yang, L.; Fang, J.; Chen, J. Tumor cell senescence response produces aggressive variants. Cell Death Discov. 2017, 3, 17049. [Google Scholar] [CrossRef] [PubMed]
  88. Tonnessen-Murray, C.A.; Frey, W.D.; Rao, S.G.; Shahbandi, A.; Ungerleider, N.A.; Olayiwola, J.O.; Murray, L.B.; Vinson, B.T.; Chrisey, D.B.; Lord, C.J.; et al. Chemotherapy-induced senescent cancer cells engulf other cells to enhance their survival. J. Cell Biol. 2019, 218, 3827–3844. [Google Scholar] [CrossRef] [PubMed]
  89. Was, H.; Czarnecka, J.; Kominek, A.; Barszcz, K.; Bernas, T.; Piwocka, K.; Kaminska, B. Some chemotherapeutics-treated colon cancer cells display a specific phenotype being a combination of stem-like and senescent cell features. Cancer Biol. Ther. 2018, 19, 63–75. [Google Scholar] [CrossRef] [PubMed]
  90. Czarnecka-Herok, J.; Sliwinska, M.A.; Herok, M.; Targonska, A.; Strzeszewska-Potyrala, A.; Bojko, A.; Wolny, A.; Mosieniak, G.; Sikora, E. Therapy-induced senescent/polyploid cancer cells undergo atypical divisions associated with altered expression of meiosis, spermatogenesis and EMT genes. Int. J. Mol. Sci. 2022, 23, 8288. [Google Scholar] [CrossRef]
  91. Kalkavan, H.; Rühl, S.; Shaw, J.J.P.; Green, D.R. Non-lethal outcomes of engaging regulated cell death pathways in cancer. Nat. Cancer 2023, 4, 795–806. [Google Scholar] [CrossRef]
  92. Pu, Y.; Li, L.; Peng, H.; Liu, L.; Heymann, D.; Robert, C.; Vallette, F.; Shen, S. Drug-tolerant persister cells in cancer: The cutting edges and future directions. Nat. Rev.Clin. Oncol. 2023, 20, 799–813. [Google Scholar] [CrossRef] [PubMed]
  93. Nano, M.; Montell, D.J. Apoptotic signaling: Beyond cell death. Semin. Cell Dev. Biol. 2024, 156, 22–34. [Google Scholar] [CrossRef]
  94. Xue, Y.; Luis, B.S.; Lane, D.P. Intratumour heterogeneity of p53 expression; causes and consequences. J. Pathol. 2019, 249, 274–285. [Google Scholar] [CrossRef] [PubMed]
  95. Mirzayans, R.; Andrais, B.; Scott, A.; Wang, Y.W.; Murray, D. Ionizing radiation-induced responses in human cells with differing TP53 status. Int. J. Mol. Sci. 2013, 14, 22409–22435. [Google Scholar] [CrossRef]
  96. Mirzayans, R.; Andrais, B.; Scott, A.; Paterson, M.C.; Murray, D. Single-cell analysis of p16INK4a and p21WAF1 expression suggests distinct mechanisms of senescence in normal human and Li-Fraumeni Syndrome fibroblasts. J. Cell. Physiol. 2010, 223, 57–67. [Google Scholar] [CrossRef]
  97. Vitale, I.; Pietrocola, F.; Guilbaud, E.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostini, M.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; et al. Apoptotic cell death in disease-Current understanding of the NCCD 2023. Cell Death Differ. 2023, 30, 1097–1154. [Google Scholar] [PubMed]
  98. Biswas, U.; Roy, R.; Ghosh, S.; Chakrabarti, G. The interplay between autophagy and apoptosis: Its implication in lung cancer and therapeutics. Cancer Lett. 2024, 585, 216662. [Google Scholar] [CrossRef] [PubMed]
  99. Kim, R.; Kin, T.; Beck, W.T. Impact of complex apoptotic signaling pathways on cancer cell sensitivity to therapy. Cancers 2024, 16, 984. [Google Scholar] [CrossRef]
  100. Newton, K.; Strasser, A.; Kayagaki, N.; Dixit, V.M. Cell death. Cell 2024, 187, 235–256. [Google Scholar] [CrossRef]
  101. Kayagaki, N.; Webster, J.D.; Newton, K. Control of cell death in health and disease. Annu. Rev. Pathol. 2024, 19, 157–180. [Google Scholar] [CrossRef] [PubMed]
  102. Tuval, A.; Strandgren, C.; Heldin, A.; Palomar-Siles, M.; Wiman, K.G.  Pharmacological reactivation of p53 in the era of precision anticancer medicine. Nat. Rev. Clin. Oncol. 2024, 21, 106–120. [Google Scholar] [CrossRef]
  103. Wang, H.; Guo, M.; Wei, H.; Chen, Y. Targeting p53 pathways: Mechanisms, structures, and advances in therapy. Signal Transduct. Target. Ther. 2023, 8, 92. [Google Scholar] [CrossRef]
  104. Levine, A.J. Targeting the p53 protein for cancer therapies: The translational impact of p53 research. Cancer Res. 2022, 82, 362–364. [Google Scholar] [CrossRef]
  105. Brown, D.W.; Beatty, P.H.; Lewis, J.D. Molecular targeting of the most functionally complex gene in precision oncology: P53. Cancers 2022, 14, 5176. [Google Scholar] [CrossRef]
  106. Murai, J.; Pommier, Y. BRCAness, homologous recombination deficiencies, and synthetic lethality. Cancer Res. 2023, 83, 1173–1174. [Google Scholar] [CrossRef]
  107. Groelly, F.J.; Fawkes, M.; Dagg, R.A.; Blackford, A.N.; Tarsounas, M. Targeting DNA damage response pathways in cancer. Nat. Rev. Cancer 2023, 23, 78–94. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Timeline of findings related to the DNA damage response that led to the somatic mutation theory of cancer. Reproduced from Huang and Zhou [15].
Figure 1. Timeline of findings related to the DNA damage response that led to the somatic mutation theory of cancer. Reproduced from Huang and Zhou [15].
Cancers 16 03106 g001
Figure 2. Fate of giant cancer cells based on discoveries reported by Walen, Rajaraman, Erenpreisa and their colleagues in the early 2000s (reviewed in [42]). These authors revealed the following sequence of events: “Cancer cells exposed to DNA-damaging agents may fail to activate early cell cycle checkpoints and thus replicate their genome on damaged templates. While some cells may die, a great number may retain viability and acquire diverse types of nuclear abnormalities including micronuclei, multiple nuclei and a highly enlarged nucleus. A proportion of multinucleated/polyploid giant cells may undergo neotic cell division, which is characterized by karyokinesis via nuclear budding followed by asymmetric cytokinesis, resulting in the generation of small mononuclear karyoplasts (also called Raju cells). These karyoplasts may resume mitotic division, and may eventually produce highly metastatic and therapy resistant descendants” [42]. Reproduced from our 2008 review [42].
Figure 2. Fate of giant cancer cells based on discoveries reported by Walen, Rajaraman, Erenpreisa and their colleagues in the early 2000s (reviewed in [42]). These authors revealed the following sequence of events: “Cancer cells exposed to DNA-damaging agents may fail to activate early cell cycle checkpoints and thus replicate their genome on damaged templates. While some cells may die, a great number may retain viability and acquire diverse types of nuclear abnormalities including micronuclei, multiple nuclei and a highly enlarged nucleus. A proportion of multinucleated/polyploid giant cells may undergo neotic cell division, which is characterized by karyokinesis via nuclear budding followed by asymmetric cytokinesis, resulting in the generation of small mononuclear karyoplasts (also called Raju cells). These karyoplasts may resume mitotic division, and may eventually produce highly metastatic and therapy resistant descendants” [42]. Reproduced from our 2008 review [42].
Cancers 16 03106 g002
Figure 3. The role of PGCCs in cancer remission (dormancy) and relapse. At the macroscopic level, (e.g., observed in live animals [82,83]), the initial cancer remission post-therapy is followed by a dormant state (also known as “senescence-like dormancy” [84] and “minimal residual disease” [85]), and eventual tumor relapse in most patients. At the microscopic level, in response to ionizing radiation [78] or chemotherapeutic drugs [82,83] (i, green circles), the majority of bulk (para-diploid) cancer cells undergo polyploidization and/or cell fusion, generating PGCCs (ii, maroon circles). During the subsequent transitional state (iii, blue circles), a proportion of PGCCs undergo depolyploidization and generate high frequencies of rapidly proliferating small-sized progeny cells that repopulate the tumor (iv, pink circles).
Figure 3. The role of PGCCs in cancer remission (dormancy) and relapse. At the macroscopic level, (e.g., observed in live animals [82,83]), the initial cancer remission post-therapy is followed by a dormant state (also known as “senescence-like dormancy” [84] and “minimal residual disease” [85]), and eventual tumor relapse in most patients. At the microscopic level, in response to ionizing radiation [78] or chemotherapeutic drugs [82,83] (i, green circles), the majority of bulk (para-diploid) cancer cells undergo polyploidization and/or cell fusion, generating PGCCs (ii, maroon circles). During the subsequent transitional state (iii, blue circles), a proportion of PGCCs undergo depolyploidization and generate high frequencies of rapidly proliferating small-sized progeny cells that repopulate the tumor (iv, pink circles).
Cancers 16 03106 g003
Figure 4. Fluorescence images showing the nuclear morphology of 2800T (Li-Fraumeni syndrome) dermal fibroblasts at different passage numbers. P, polyploid/multinucleated giant cells. Scale bars, 50 µm. Reproduced from Mirzayans et al. [96] with permission.
Figure 4. Fluorescence images showing the nuclear morphology of 2800T (Li-Fraumeni syndrome) dermal fibroblasts at different passage numbers. P, polyploid/multinucleated giant cells. Scale bars, 50 µm. Reproduced from Mirzayans et al. [96] with permission.
Cancers 16 03106 g004
Table 1. Clinical and molecular characteristics of the indicated human cancer-prone disorders. For details, see [43].
Table 1. Clinical and molecular characteristics of the indicated human cancer-prone disorders. For details, see [43].
DisorderMode of TransmissionDefective ProteinDefective Function
Xeroderma pigmentosumAutosomal recessiveXPA through XPG,
DNA polymerase η
Nucleotide excision repair (XPA through XPG), postreplication repair (XPV)
Ataxia telangiectasiaAutosomal recessiveATMATM signaling
Li-Fraumeni syndromeAutosomal dominantp53; Chk2p53/Chk2 signaling
Nijmegen breakage syndromeAutosomal recessiveNBS1DSB repair
Werner syndromeAutosomal recessiveWRNDNA helicase
Bloom syndromeAutosomal recessiveBSDNA helicase
Rothmund-Thompson syndromeAutosomal recessiveRTSDNA helicase
Fanconi anemiaAutosomal recessiveFANCA, B, C, D1, D2, E, F, G, I, J, L and MDNA helicase,
DNA cross-link repair
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mirzayans, R.; Murray, D. Amitotic Cell Division, Malignancy, and Resistance to Anticancer Agents: A Tribute to Drs. Walen and Rajaraman. Cancers 2024, 16, 3106. https://doi.org/10.3390/cancers16173106

AMA Style

Mirzayans R, Murray D. Amitotic Cell Division, Malignancy, and Resistance to Anticancer Agents: A Tribute to Drs. Walen and Rajaraman. Cancers. 2024; 16(17):3106. https://doi.org/10.3390/cancers16173106

Chicago/Turabian Style

Mirzayans, Razmik, and David Murray. 2024. "Amitotic Cell Division, Malignancy, and Resistance to Anticancer Agents: A Tribute to Drs. Walen and Rajaraman" Cancers 16, no. 17: 3106. https://doi.org/10.3390/cancers16173106

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop