Abstract
Those diagnosed with osteosarcoma today receive the same therapy that others received over the last four decades. Extensive efforts to identify more effective or less toxic regimens have proved disappointing. As we enter a post-genomic era, now recognizing osteosarcoma not as a cancer of mutations but as one defined by p53 loss, chromosomal complexity, copy number alteration, and profound heterogeneity, emerging threads of discovery leave many hopeful that an improving understanding of biology will drive discoveries that improve clinical care. Under the organization of the Bone Tumor Biology Committee of the Children’s Oncology Group, a team of clinicians and scientists sought to define the state-of-the-science and to identify questions that, if answered, have the greatest potential to drive fundamental clinical advances.
Having discussed these questions in a series of meetings, each led by invited experts, we distilled these conversations into a series of seven Provocative Questions. These include questions about the molecular events that trigger oncogenesis, the genomic and epigenomic drivers of disease, the biology of lung metastasis, research models that best predict clinical outcomes, and processes for translating findings into clinical trials.
Here, we briefly present each Provocative Question, reviewing the current scientific evidence, noting the immediate opportunities, and speculating on the impact that answered questions might have on the field. We do so with an intent to provide a framework around which investigators can build programs and collaborations to tackle the hardest problems and to establish research priorities for those developing policies and providing funding.
Keywords: osteosarcoma, pediatric oncology, translational biology, sarcoma, program development
Precis:
Scientific advances have significantly improved our understanding of osteosarcoma but gaps in our knowledge have impaired our ability to translate new findings into new cures. Here, we present a series of questions that we believe represent the most important and most immediate unsolved problems which, if solved, might change the way we think about and treat this disease.
Introduction
Osteosarcoma (OS) is an aggressive bone malignancy that primarily affects adolescents and young adults. The standard-of-care regimen used today was first introduced in the late 1970s1 and remains largely unaltered despite numerous efforts to improve outcomes2,3. Patients who present with localized disease today still face 5-year overall survival rates of less than 70%; less than 20% of those who develop metastatic disease or recurrence survive beyond 3 years4.
Recent experience suggests that transformative advances in the treatment of this disease will not likely come from intensification of antineoplastic chemotherapy5, further focusing efforts on developing truly novel approaches. A fresh surge of scientific discovery has fueled an evolving appreciation for the complex biology of this malignancy, giving life to several ongoing clinical and basic scientific efforts that seemingly have potential to impact patient outcomes.
In an effort to create an environment that could foster collaborative thought and discussion around these opportunities, the Bone Tumor Committee of the Children’s Oncology Group (COG) launched a monthly web-based conference for basic and translational scientists interested in OS research. The charge of the reformed Osteosarcoma Biology Committee was to bring together scientists committed to sharing their work, identify significant hurdles that preclude deeper understanding of OS biology, and establish new research collaborations. Participants were identified by personal referral of existing members of the COG Bone Tumor Committee, requiring only interest and a willingness to actively participate. Nearly 50 individuals were initially invited and participation continues to grow. Monthly calls routinely have more than 30 worldwide participants.
In our inaugural meeting, the group galvanized around a plan to use this forum to identify and explore key gaps in our understanding of OS biology. By methodically reviewing the most current and potentially-impactful opportunities for discovery science and clinical translation, we hoped to create a blueprint that could shape the priorities of scientists, foundations, advocacy groups, and funding agencies around common goals.
To this end, we asked members of the committee to submit one or more candidate Osteosarcoma Provocative Question that would encapsulate specific challenges to our basic, translational, and clinical understanding of the disease. We consolidated submissions into a series of OSPQs (Box 1), then systematically reviewed the state-of-the-art and current opportunities as a group in our open meetings. We invited experts (both from within and from beyond our group) who could proficiently address each question, asking them to summarize the most current science, reserving the majority of each session for active discussion. The results of these discussions are summarized here as a series of OSPQs, refined by those discussions, and presented in a format consistent with the National Cancer Institute’s Provocative Questions Initiative6.
Box 1. Osteosarcoma Provocative Questions (OSPQs).
OSPQ1: What are the disease initiating events in osteosarcoma and how do those events lead to genomic complexity and cellular heterogeneity?
OSPQ2: What can epigenetic profiling tell us about osteosarcoma?
OSPQ3: What can we learn from studying the inherited cancer susceptibility syndromes that increase the risk of osteosarcoma?
OSPQ4: Which research models best represent the biology of osteosarcoma?
OSPQ5: What biological mechanisms mediate osteosarcoma lung metastasis?
OSPQ6. What factors limit the efficacy of immuno-oncology approaches in osteosarcoma?
OSPQ7: Can emerging clinical trial designs compliment and accelerate the development of novel approaches to osteosarcoma?
OSPQ1: What are the disease-initiating events in osteosarcoma and how do those events lead to genomic complexity and cellular heterogeneity?
Intent.
Despite multiple intensive efforts to genomically characterize OS, a common inciting event remains elusive. While bi-allelic loss of TP53 appears to be a near universal requirement for the development of OS, it remains unclear whether loss of TP53 itself is a sufficient oncogenic event. One major challenge to interpreting the results of large-scale genomic efforts is the seemingly stochastic complexity and heterogeneity encountered in OS tumors. Which of the identified alterations represent true drivers of malignancy? Which can be targeted therapeutically? Which features provide prognostic or diagnostic value?
Background.
Most OS tumors harbor inactivating TP53 mutations or amplifications of MDM2/47,8. The frequency of TP53 mutations was previously underestimated until intronic rearrangements, largely in intron 1, were recognized as a frequent mechanism of somatic TP53 loss9,10. The high allelic fraction of TP53 loss suggests that mutations in TP53 occur as a very early event in oncogenesis. Several studies, including a large meta-analysis have associated specific genetic TP53 alterations with survival in patients with OS11, suggesting mutation-specific phenotypes. Many tumors also bear RB mutations and, less commonly, mutations in ATRX, cell cycle proteins, and the PTEN/PI3K pathway9.
Beyond this small set of mutations, the most common characteristic of OS is structural complexity through chromosomal rearrangements, copy number variation, kataegis, and chromothripsis, which occur broadly throughout the genome9,12. Widespread corruption of chromosomal structure may produce progeny with divergent patterns of copy number variation that undergo serial selection for growth advantage. Indeed, recent work has shown that a limited set of copy number patterns can group OS tumors into subtypes that may predict response to certain targeted agents13. These patterns, however, explain only a small number of events and the degree of cell-to-cell heterogeneity within a tumor remains unknown, though chromosomal heterogeneity could establish mechanisms for diversity that would facilitate adaptation to cellular stresses, such as chemotherapy.
Surprisingly, OS lacks common or recurrent changes in DNA repair pathway genes. Rather than accumulating this complexity over time, mathematical modeling of osteosarcomagenesis based on a cell cycle time scale suggests that the critical genetic events leading to malignancy occur abruptly within a single cell cycle14,15. In fact, unchecked cell cycle progression due to TP53 loss may be sufficient to initiate oncogenic transformation in the correct cell-of-origin and microenvironmental context.
Feasibility.
Numerous pan-cancer genomic studies have elucidated the role of single-nucleotide variants and small focal copy-number alterations in OS biology. Widespread chromosomal copy-number changes, chromothripsis, kataegis, and aneuploidy have been clearly identified as characteristic features of OS, though their role in the oncogenic process remains largely unknown. With several published and unpublished sequencing projects now completed9,12, an effort to combine all available data into a harmonized cohort would be better powered to identify recurrent somatic events and subtypes of OS. Improving the availability of paired samples should allow more sophisticated comparison of primary, recurrent, and metastatic lesions. Emerging technologies, such as single-cell sequencing approaches, may help distinguish genomic complexity from cell-to-cell heterogeneity. Furthermore, integration of epigenomic and proteomic data may facilitate the interpretation of genomic structural variants and the elucidation of their biological effects. Finally, accurate in vitro and in vivo models, as described in other sections below, should help identify targetable dependencies engendered by osteosarcoma’s unique genomic profile.
Implications of success.
A mechanistic understanding of the biological processes that drive osteosarcomagenesis and malignant progression would move the field forward in a number of substantive ways. First, a detailed grasp of the biological mechanisms that lead to malignancy in this disease would facilitate assessment of the relevance of different models of OS and may help produce models more faithful to the human disease. Second, separating the driver genetic changes from the genetic noise of passenger variants would help focus research efforts on relevant biology and may reveal specific therapeutic vulnerabilities. Indeed, if emerging ideas hold true, which hypothesize a common proximal event that sets the stage for the selection of specific patterns of copy number variation that produce growth advantage, the refinement and study of distinct subtypes could represent a paradigm shift in the way we think about treating this disease.
OSPQ2: What can epigenetic profiling tell us about osteosarcoma?
Intent.
While widespread structural variations leading to copy number gains and losses characterize the genetic landscape9,12,23, the role of epigenetic dysregulation in osteosarcomagenesis and malignant progression remain less explored. What specific epigenetic mechanisms contribute to OS? How does the epigenetic landscape of OS compare to the putative cells of origin? Are epigenetic patterns common across the majority of osteosarcomas or heterogeneous across tumors? Are these changes heterogeneous within a single tumor site? What epigenetic changes drive progression, resistance, and metastasis? Do changes resulting from gains or losses of proteins that function within epigenetic regulatory complexes influence OS cell biology?
Background.
Increasingly, regulatory or functional changes in DNA methylation, enhancers, superenhancers and histone state have been recognized as primary drivers of pediatric cancers. While studies reporting the disease-specific epigenetic landscape of OS have not been reported, both the TARGET and Heidleberg24 genomic efforts are generating DNA methylation data in large clinically-annotated sample sets. Preliminary reports suggest that the methylation signatures in each independent dataset do not have a clear underlying oncogenetic correlation. A few osteosarcomas have methylation patterns outside the primary clusters that coincide with genetic aberrations in epigenetic regulatory genes.
While mounting evidence suggests surprising stability of the methylome from primary to metastatic sites, focused preclinical models have recently demonstrated that OS cells possess an ability to alter enhancer profiles and patterns of transcription that enable cells to metastasize and proliferate in the lung microenvironment25,26. Leveraging these epigenetic patterns to identify common state-specific treatment vulnerabilities may enable the development of therapies tailored to metastatic lesions25.
Feasibility.
Epigenetic lines of inquiry have potential to enhance our understanding of OS development, plasticity, mechanisms of metastasis, and the biology of treatment resistance. Since the underlying features that allow for numerous structural variations in OS remain unknown, it is possible that epigenetic mechanisms are involved from very early stages of osteosarcomagenesis. Methylation patterns may provide insights into the cell of origin. Targeted perturbations of specific enhancer elements may reduce metastatic potential of OS cells.
Ultimately, analyses that integrate the latest genomic, proteomic and epigenetic information will likely be the most informative. Additional insights can emerge when such analyses are enhanced with matched metastatic or relapse samples and robust clinical data. Future banking strategies for patients with OS undergoing surgical resections or biopsies should recognize the value that well-annotated samples, properly handled and preserved, sufficient for performing global analyses, might have27.
Implications of success.
Remaining questions about OS, including mechanisms of metastasis, patient-to-patient and cell-to-cell heterogeneity, pervasive structural genetic changes, and the elusive cell of origin might be answered through a more detailed understanding of epigenetics in OS. Therapeutic avenues for targeting epigenomic dependencies are in early stages of development, but have promise for clinical translation.
OSPQ3: What can we learn from studying the inherited cancer susceptibility syndromes that increase the risk of osteosarcoma?
Intent.
Osteosarcoma is the most common childhood cancer associated with congenital loss-of-function mutations in both TP53 (Li-Fraumeni Syndrome) and RB1, the two genes recurrently mutated in patients with the disease. However, increased incidence of OS is not limited to these predisposition syndromes. Indeed, germline mutations in other genes can confer even greater risk for OS. Given the poorly understood molecular origins of OS, patients carrying germline mutations that predispose to the disease present unique opportunities to learn about osteosarcomagenesis. It would be prudent to ask what might be learned from these patients and how this might help them. What subsequent molecular alterations lead to malignant progression in these patients? Why do non-TP53/non-RB1 mutations also lead to OS? Are these the same diseases? Do different types of TP53 mutations alter the clinical phenotype of the disease?
Background.
Several cancer predisposition syndromes are associated with an increased risk of OS. The highest and most specific risk for OS occurs in patients with Rothmund-Thomson Syndrome (RTS)28, an autosomal recessive disorder caused by germline mutations in the DNA helicase gene, RECQL4. (The incidence of OS in RTS patients is approximately 30%29.) Syndromes defined by mutations in other DNA helicase genes also confer an increased risk of OS, including Bloom, Werner, and RAPADILINO syndromes30. OS is the most common cancer of childhood associated with Li-Fraumeni Syndrome, caused by germline TP53 mutations, occurring with a cumulative incidence of 5–11%31–33 in these patients. Indeed, it is estimated that approximately 10% of all patients with OS aged less than 30 years have germline TP53 mutations34.
Feasibility.
The major cancer susceptibility genes associated with OS are known. Animal models are available for the most common gene mutations (i.e. TP53 and RECQL4). Investigators should be able to delve more deeply into the early events and networks that are important in OS development and to study how those events interact with or are influenced by other underlying germline polymorphisms. In addition, studies comparing patterns of somatic mutations and epigenomic profiles of tumors from patients with and without underlying germline cancer predisposition syndromes could be performed with existing technologies to understand the differences in tumor biology in these cohorts. Evaluation of the clinical significance of germline mutations on OS outcomes will require collaborative efforts to combine data from single institutions and cooperative groups, such as the Children’s Oncology Group. Given that upfront treatment of OS in the United States is largely universal with methotrexate, doxorubicin, and cisplatin (MAP), comparisons across institutions should be feasible if a coordinated effort can be organized.
Implications of success.
The successful results of this research will define how genetic factors interact to augment the action of cancer susceptibility genes to promote OS development. Moreover, molecular investigations into the interactions of those genes and exposures may identify networks and pathways that are critical to the development of this disease. Finally, a coordinated effort to examine the outcomes of OS patients with inherited genetic cancer susceptibility may broadly inform investigators on the mechanisms that promote or inhibit clinically relevant mechanisms such as metastasis and chemosensitivity.
OSPQ4: Which research models best represent the biology of osteosarcoma?
Intent.
While researchers continue to utilize, develop, and characterize an ever-growing collection of disease models, discerning which models best represent the biology of human OS remains difficult. The translational value of scientific discoveries hinges on the answers to a series of fundamental questions that, in many cases, remain difficult to answer: How closely do cellular and animal models of OS recapitulate the biologic and ‘omic features (genomic, transcriptomic, epigenomic, etc.) of the human disease? How much (and when) does the immune and stromal environment contribute to the interpretation of results from each model? Does exposure of the model to agents with known activity in the human disease induce a similar response in the model? And, most importantly, does response in the model to novel therapies predict responses in patients with OS?
Background.
To study OS at both a basic and a preclinical level, investigators rely on a broad range of models, including established cell lines39, patient-derived cell cultures, patient-derived xenografts13,40–42, cell line xenografts, and genetically engineered mice43–45. Pet dogs that spontaneously develop OS represent a parallel patient population that presents unique opportunities for generating preclinical human data43,46. These models differ widely with respect to their distance from the patient, their ease and cost of use, and the fidelity with which they recapitulate different elements of the human disease. In many cases, these determinations can be difficult to make due to a lack of existing data.
One significant barrier to determining the predictability of existing models is the lack of recent clinical trials with positive results, which makes it difficult to identify predictors of efficacy. Negative clinical trials give the impression that preclinical models were not accurate in predicting response. However, others might argue that this results from setting an unreasonably low bar for calling a preclinical study “positive”.
As the number of biological leads expands and our approach to treatment becomes more sophisticated through increasingly complex precision oncology frameworks, immune- and stromal-targeting strategies, and combinatorial regimens, our need to understand which models faithfully recapitulate specific elements of OS biology becomes more urgent. From a basic science perspective, our incomplete understanding of the fundamental biology underlying osteosarcomagenesis44,47 and most aspects of malignant progression limits our ability to discern the value of numerous distinct models of disease.
Pet dogs frequently develop OS, which bears striking clinical, genetic, and biologic similarity to the human malignancy and almost always leads to metastatic pulmonary disease46,48,49. The spontaneous nature of this disease, the shared environmental context, the shorter natural history of the dog disease, and the increasing capacity of collaborative clinical trials groups within the veterinary community make this a primary early target of comparative oncology efforts50,51. The canine patient may become a valuable tool for vetting preclinical candidates, for answering important pharmacodynamic questions, and for the parallel evaluation of biologic responses to the perturbations induced by candidate interventions.
Feasibility.
While genomic and other high dimensional characterizations of human OS tumors and models have been completed, only very limited ‘omic comparisons of cell lines, patient-derived xenografts (PDXs), and genetically-engineered mouse models (GEMs) to primary human samples have been performed. Likewise, although comparative work in canine models suggests marked commonalities between human and canine OS48,52, evaluation using modern techniques to compare this to the human disease still warrants attention. Such research could help scientists select models that best represent aspects of OS particularly suited to the testing of novel therapeutics, also leading to identification of biomarkers and precision implementation of clinical trials. Promising candidates in the mouse could be rapidly vetted in canine trials of new OS-targeting agents.
Using readily available models and techniques, several coordinated efforts could be considered to address this question. These might include:
Profiling of GEMs and PDXs to reveal their genomic, epigenomic, and proteomic features. This data could be superimposed on the human data to “bin” models into appropriate subclasses (or to deem them either broadly applicable or not representative of human OS).
Comprehensive profiling of canine tumors performed in parallel with analysis of human data to determine whether similar subtypes of OS are present or absent in dogs.
Making the broad range of already developed OS models readily accessible to scientists, including the relevant characterization data associated with each model.
Development of a more coordinated enterprise that integrates and aligns the work of veterinary clinical trials groups with their pediatric counterparts. Opportunities exist to coordinate the work of canine clinical trials consortia (such as the Comparative Oncology Trials Consortium and the Canine Immunotherapy Trials and Correlative Studies Group, which was created through a recent series of Moonshot grants) with that of their human counterparts (COG and other clinical trials consortia) simply by including members of both groups in their respective planning and development meetings.
Until we have clinical results that can clarify the predictability of our models, the relative value of preclinical data remains difficult to assess. A formalized scoring system for prioritizing and valuating preclinical data exists53, though few have utilized this system in reporting preclinical results. Improving dialogue between trialists and the research community could convey realistic expectations to the clinicians, relay important pharmacokinetic considerations, assure that optimizations to scheduling or dosing are reflected in trial design, and facilitate collection of appropriate samples for correlative study. In the short term, a retrospective analyses of the predictive performance of preclinical models relative to clinical trial outcomes could help identify aspects of experiments that are more predictive than others.
Implications of success.
As we enter an era where clinical decisions and preclinical development plans have become increasingly driven by high-level characterization of tumors and refinement of diagnostic criteria (which divide tumors into increasingly small but specific groups), the importance of understanding how our preclinical models recapitulate this biology cannot be overstated. Work done to determine which results from which preclinical models will accurately predict results in patients with OS has enormous potential to speed therapeutic development.
OSPQ5: What biological mechanisms mediate osteosarcoma lung metastasis?
Intent.
Metastatic dissemination and distant recurrence occurs primarily in the lung54. This natural history suggests that elucidating the mechanisms that facilitate lung metastasis might identify clinically-relevant targetable vulnerabilities. Despite the seemingly obvious potential of such strategies, we currently understand very little about the biology that drives lung colonization. What specific adaptations or intracellular signals intrinsic to certain tumor cells make OS cells survive and grow in the lung? What lung-derived signals activate survival pathways? Does lung-induced dormancy protect disseminated tumor cells from chemotherapy or mediate late metastatic relapse?
Background.
Metastatic disease is the most critical clinical factor that influences malignant progression and mortality in OS53. Yet we know very little about the biology underlying this metastatic process. We do know that in the early stages of metastatic lung colonization, disseminated cancer cells experience a variety of cellular stresses (e.g. redox/endoplasmic reticulum stress) that threaten their survival in the distant microenvironment19,55,56. Metastatic inefficiency arises from the inability of most cells to survive the stresses involved with initial arrest within a distant tissue57–59. Animal studies using well-characterized xenograft models of metastatic OS60 have identified a number of molecular pathways important for metastatic progression18,19,61–64. However, the clinical relevance of these potential vulnerabilities has yet to be determined in patients.
Dormancy of disseminated cells within the lung could explain the most common pattern of metastasis—late recurrence. Several groups have identified potential drivers of OS dormancy65–67, including suggestions that tumor-lung interactions might support the dormancy state, though this remains a relatively unexplored field. Additional studies are needed to validate these findings.
Feasibility.
Since lung metastasis is a dynamic, multi-step process that involves interactions between OS cells, extracellular matrix, and lung parenchymal cells, model systems must facilitate the responses of OS cells to pharmacologic or genetic manipulation within the lung microenvironment. Several existing models are available that could help our understanding of lung-specific responses to perturbagens. For example, the ex vivo pulmonary metastasis assay (PuMA)68–70 permits researchers to directly observe OS cell growth within the lung microenvironment while precisely controlling any number of experimental conditions. Many xenograft and GEM models of OS faithfully recapitulate patterns of metastasis and tumor-lung interactions observed in the human condition (discussed in OSPQ4). Intravital imaging techniques also now allow for high-resolution, longitudinal imaging of emerging metastases in context58,71. Single-cell whole-genome analyses offer enormous promise for furthering our mechanistic understanding of tumor-host interactions within the metastatic niche.
Implications of success.
Lung metastasis defines prognosis in patients with OS. A therapy that effectively prevents the emergence of lung metastasis could potentially save as many as 70% of the lives currently lost to the disease53. Therapies which render metastatic disease treatable could have even greater impact. Indeed, targeting metastatic disease has been identified as a research priority by a diverse group of experts53, and well-vetted approaches that target prevention or treatment of metastasis would likely be prioritized for clinical evaluation.
OSPQ6. What factors limit the efficacy of immuno-oncology approaches in osteosarcoma?
Intent.
Interaction of tumor and host immune environment has long been believed to be a critical aspect of tumor survival in malignancies. Despite recent success of immune therapies in several tumor histologies and identification of some key factors that may predict response such as a high tumor mutational burden or DNA mismatch repair defects72–74, the relationship between tumor and host immune microenvironment, their interactions and opportunities for exploitation with immune-mediated therapies remain poorly defined in OS. The key questions to answer in this context in OS include: What is known currently about the tumor immune profile? Are there differences among host immune profiles of patients with OS as compared to healthy controls and do these differences matter? Do OS primary and/or metastatic tumors have the right milieu for immune therapies to be effective? What can we do to make immune therapies effective in these tumors and patients? What models can we use to study these questions?
Background.
OS has been considered to be immunogenic and amenable to immune therapies since the early 19th century when William Coley, considered to be the “Father of Immunotherapy”, observed responses after injecting patients with bone and soft tissue sarcomas with streptococcal-derived toxins (Coley’s toxins)75. In recent years, the recognition of extreme genomic complexity of OS76,77 has led many to believe that a higher tumor mutational burden and neo-epitope antigen generation should suggest response to immune therapies such as immune checkpoint inhibition. However, while OS tumors do exhibit higher mutational burden than many pediatric cancers, this is still much lower than a majority of adult cancers24.
Other evidence points to a potential role of the immune microenvironment in OS progression. Several studies suggest that infiltration of tumors with cytotoxic T cells as well as tumor associated macrophages predicts improved survival in patients with OS78–80. Further, expression of PD-1 on tumor infiltrating lymphocytes and its ligand PD-L1 on tumor cells has been shown both in primary OS tumors as well as lung metastases, suggesting that this pathway might play a role in tumor microenvironment78,81. When compared to healthy controls, increased expression of CTLA4 on peripheral T cells and an increased ratio of immune suppressive peripheral monocytes in patients with OS may suggest a profoundly immunosuppressive environment within tumors79. Preclinical studies in immunocompetent mouse models suggest that blockade of immune checkpoint pathways alone or in combination with chemotherapy (trabectedin) or radiation can reduce both primary and metastatic tumor burden in bone and soft tissue sarcomas81–84.
Despite this preclinical evidence of immune checkpoint inhibitors activity, the response to these agents in clinical trials has been underwhelming. In SARC028 study, only one of the 22 patients with OS had an objective response to single agent pembrolizumab85. In the pediatric trial of nivolumab alone or in combination with ipilumumab, no objective responses were seen in patients with OS. It is clear from these results that more work needs to be done to understand the discrepancy between preclinical and clinical outcomes and to establish novel ways to improve responses to immune therapies in this disease.
Feasibility.
While initial clinical trials have been far from promising, strong preclinical data suggests an untapped potential for harnessing the immune system against OS. With an abundance of new genomic and transcriptomic data now available on OS, it is imperative that there is close collaboration between cancer immunologists and genomicists to determine which targetable immune regulatory proteins, cell therapies and combination treatments have the most potential for this disease. Our current understanding of the tumor/immune microenvironment of OS appears weak and warrants intensive biological study.
Identifying appropriate model systems for these experiments presents a particular challenge. Rodent models of disease that faithfully mimic the human immune microenvironment do not currently exist, although efforts to develop humanized immune competent mice are ongoing86. Canine models and veterinary clinical trials may meet some of these needs. Efforts have been underway within Children’s Oncology Group to develop clinical trials of immune checkpoint inhibitors in combination with chemoradiotherapy based on intriguing preliminary evidence suggesting that radiation might immunosensitize these tumors83. Other efforts to identify novel therapies that effectively modulate the immune response to OS, such as targeting IDO, B7-H3 or certain chemokines (CXCL12) are also under consideration. If designed with the proper integrated correlative biology experiments, these trials could significantly enrich our understanding of the role of the immunity in this disease.
Implications of Success.
Advances in OS therapy have stalled over many decades. Comprehensive genomic and transcriptomic profiling has thus far failed to identify easily druggable targets. Given the recent successes in other cancer types, immune therapies should be explored to determine their potential for efficacy in OS. Successful recognition of tumor and host immune environment and of barriers that prevent anti-tumor immune effectors from productively eliminating tumor cells could lead to rationally-designed immune therapy combinations in OS.
OSPQ7: Can emerging clinical trial designs compliment and accelerate the development of novel approaches to osteosarcoma?
Intent.
Most of the advances made in the treatment of OS developed empirically, during a time when few systemic options existed and little data was needed to support investment in a clinical trial. Since that time, the demand for pre-clinical data to justify the initiation of a clinical trial has grown, even as the parameters for what constitutes sufficient pre-clinical evidence remains undefined (OSPQ4). However, in a disease where definitive, prospective clinical trials often require more than 10 years from conception to initial results87, delays in initiating trials based on good clinical intuition could result in decades of stagnation in how patients with OS are treated. Beyond identification of a potentially efficacious therapy, another opportunity cost of not having active clinical trials is reduced ability to collect and analyze biological specimens towards biomarker discovery and improved understanding of biology. Patients have been eager to test novel therapies, even when those studies are unlikely to benefit them personally88,89. Fortunately, newer phase II study designs facilitate trials with smaller patient cohorts, which could create more opportunities to study novel therapies for patients with recurrent disease90. With this, can trialists and researchers leverage smaller, leaner trial designs to not only vet treatments with strong preclinical justification, but to accelerate the development of newer ideas in a more integrated preclinical/early clinical environment? Should the go/no-go threshold be influenced by the lack of other therapeutic options for patients, such as off-label agents or competing trials? How can alternative mechanisms of efficacy such as inhibition of metastases, rather than cytotoxic effects, be tested and translated? Is it ever acceptable to test ideas directly in patients without preclinical data? Similar to questions asked for preclinical models, does response in patients with relapsed disease predict activity in patients with newly diagnosed disease?
Background.
Accepting that additional intensification of cytotoxic agents will not improve outcomes in patients with OS2,91, investigators have turned to an ever-lengthening list of tyrosine kinase inhibitors, immunotherapies, and targeted agents with increasingly diverse and novel mechanisms of action as potential treatments for OS. This creates a need for smaller clinical trials that can vet more potential therapies more quickly. Such efforts have long been hampered by the lack of an acceptable historical control. Fortunately, a dedicated analysis of past trials has been completed and accepted as a historical control that trialists can use to power and evaluate future clinical trials in patients with relapse OS90. This transition to a more relevant, easily measured efficacy threshold circumvents the problems inherent to the application of more widely used response measures inherent to OS92. Thus there are clear efficacy bars for phase II trials. Using these standards, trials can be conducted in under 3 years93,94.
Other emerging lines of work could significantly affect how we conduct clinical trials in osteosarcoma. For instance, Bayesian trial designs might have particular value in rapidly evaluating multiple treatments for rare tumors like osteosarcoma95. Also, if OS truly represents a collection of biologically-distinct clinical entities (OSPQ113), the use of biomarkers to classify individuals into smaller, biologically-related groups would dramatically affect our approach to conducting clinical trials.
Feasibility.
With no first-line therapeutic trial currently open and with a relatively small number of phase 2 trials available for relapsed patients, the number of children with osteosarcoma enrolled in NCI-sponsored clinical trials has decreased significantly compared to when frontline trials have been enrolling. This has created a situation with more patients interested in enrolling in clinical trials than there are trials available, evidenced by the rapid accrual of recent trials that have far exceeded enrollment expectations93,94. While numerous therapeutic strategies have some preclinical evidence supporting further investigation, very few have a level of evidence that would meet traditional standards for justifying a clinical trial. Many of these concepts invoke treatment strategies already employed in other diseases and with known safety profiles, lowering the potential risks of use in this population.
Ideally, OS research would use a more integrated approach, testing novel regimens across collections of diverse models, with laboratory experiments that educate human clinical trials96 and canine clinical trials48,97 and vice-versa. Each study, each experiment designed to answer important questions pertinent to that approach, all informing the ongoing development of future human studies by optimizing dose, schedule, biomarkers for inclusion/exclusion, and the thoughtful implementation of meaningful correlative studies98.
Implications of success.
Establishing a clear threshold of preclinical data to justify a clinical trial would help those designing and planning experiments in OS. Widespread agreement concerning phase 2 efficacy thresholds should expand the number of relapse trials available by creating a transparent set of rules that facilitate study design and appropriate powering. While these represent an incredibly valuable first step, consideration should also be given to other clinical trials approaches that might also accelerate clinical trials timelines. Tighter integration of our preclinical research enterprise with our clinical trials networks could improve our ability to determine the positive or negative predictive value of preclinical data and accelerate solutions that will improve patients’ chances of responding when enrolling on clinical trials.
Conclusions
While some say that history should temper the optimism of those who look for science to impact outcomes in patients with OS in the near future, recently converging efforts have matured in ways that dramatically improve our understanding of osteosarcoma’s underlying biology. This acceleration of scientific discovery has the potential to drive a revolution in the care of affected patients. Our rapidly improving understanding of the genetic events that drive disease at both the genetic and epigenetic level, informed by predisposition science, coupled with studies that describe the biology of progression with increasing detail have set the stage. Growing collections of PDXs, cell lines, and banked human tissues, and an increasingly agile multi-institutional veterinary clinical trials groups should enable scientific progress with increasing efficiency. Human clinical trials consortia have begun to explore new models for evaluating candidate therapies, and immunotherapeutic strategies have become increasingly sophisticated in ways that may have promise for patients with OS. We believe that opportunities to translate scientific knowledge into clinical impact have never been greater.
However, much work remains. Our efforts highlight a number of critical gaps that could prevent us from translating our new findings into new cures for patients with OS. In organizing these gaps into a set of OSPQs, we highlight here the clear need for investment in all three phases of research: basic, translational, and clinical. Two recurring major themes ran throughout the discussions that fueled the creation of this list of OSPQs: First, while our understanding of the basic biologic mechanisms of OS genesis and progression has improved, there remain a large number of unanswered fundamental questions. Second, better characterization of preclinical models and a better understanding of their utility for predicting outcomes in patients would significantly improve our ability to validate new therapeutic candidates as they emerge. We hope that this codification of the most prominent challenges will serve to organize the efforts of government, philanthropic, and advocacy groups to facilitate meaningful science and will promote interaction within our scientific communities in ways that leverage our collective resources toward achieving a common goal—to offer our patients something far better than we can now.
We extend an open invitation to those physicians and scientists with interest in participating in our monthly calls. To initiate participation, please email the corresponding author.
Funding:
This work was made possible through an NCTN Operations Center Grant, U10CA180886.
Footnotes
Conflict of interest disclosures:
The authors declare that they have no conflicts of interest in regard to this manuscript.
References
- 1.Winkler K, Beron G, Delling G, et al. Neoadjuvant chemotherapy of osteosarcoma: results of a randomized cooperative trial (COSS-82) with salvage chemotherapy based on histological tumor response. J Clin Oncol. 1988;6(2):329–337. [DOI] [PubMed] [Google Scholar]
- 2.Marina NM, Smeland S, Bielack SS, et al. Comparison of MAPIE versus MAP in patients with a poor response to preoperative chemotherapy for newly diagnosed high-grade osteosarcoma (EURAMOS-1): an open-label, international, randomised controlled trial. Lancet Oncol. 2016. doi: 10.1016/S1470-2045(16)30214-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bielack SS, Werner M, Tunn PU, et al. Methotrexate, doxorubicin, and cisplatin (MAP) plus maintenance pegylated interferon alfa-2b versus MAP alone in patients with resectable high-grade osteosarcoma and good histologic response to preoperative MAP: First results of the EURAMOS-1 good respons. J Clin Oncol. 2015;33(20):2279–2287. doi: 10.1200/JCO.2014.60.0734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Gorlick R, Janeway K, Lessnick S, Randall RL, Marina N. Children’s Oncology Group’s 2013 blueprint for research: bone tumors. Pediatr Blood Cancer. 2013;60(6):1009–1015. doi: 10.1002/pbc.24429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kleinerman E Maximum benefit of chemotherapy for osteosarcoma achieved-what are the next steps? Lancet Oncol. 2016;17(10):1340–1342. doi: 10.1016/S1470-2045(16)30270-4. [DOI] [PubMed] [Google Scholar]
- 6.Berny-Lang MA, Greenspan EJ. NCI’s Provocative Questions Initiative In: Cancer Research. Vol 74 American Association for Cancer Research; 2014:3500–3500. doi: 10.1158/1538-7445.AM2014-3500. [DOI] [Google Scholar]
- 7.Chauhan KM, Ramakrishnan G, Kollareddy M, Martinez LA. Characterization of cancer-associated missense mutations in MDM2. Mol Cell Oncol. 2016;3(2):e1125986. doi: 10.1080/23723556.2015.1125986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Miller CW, Aslo A, Won A, Tan M, Lampkin B, Koeffler HP. Alterations of the p53, Rb and MDM2 genes in osteosarcoma. J Cancer Res Clin Oncol. 1996;122(9):559–565. [DOI] [PubMed] [Google Scholar]
- 9.Chen X, Bahrami A, Pappo A, et al. Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep. 2014;7(1):104–112. doi: 10.1016/j.celrep.2014.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ribi S, Baumhoer D, Lee K, et al. TP53 intron 1 hotspot rearrangements are specific to sporadic osteosarcoma and can cause Li-Fraumeni syndrome. Oncotarget. 2015;6(10):7727–7740. doi: 10.18632/oncotarget.3115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chen Z, Guo J, Zhang K, Guo Y. TP53 Mutations and Survival in Osteosarcoma Patients: A Meta-Analysis of Published Data. Dis Markers. 2016;2016:4639575. doi: 10.1155/2016/4639575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Perry JA, Kiezun A, Tonzi P, et al. Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma. Proc Natl Acad Sci U S A. 2014;111(51):E5564–73. doi: 10.1073/pnas.1419260111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sayles LC, Breese MR, Koehne AL, et al. Genome-Informed Targeted Therapy for Osteosarcoma. Cancer Discov. September 2018:CD-17–1152. doi: 10.1158/2159-8290.CD-17-1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ma X, Liu Y, Liu Y, et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature. 2018;555(7696):371–376. doi: 10.1038/nature25795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bilke S, Meltzer PS. Abstract LB-291: Environment or accident? A more stringent bound on environmental contributions to cancerogenesis. Cancer Res. 2016;76(14 Supplement):LB-291–LB-291. doi: 10.1158/1538-7445.AM2016-LB-291. [DOI] [Google Scholar]
- 16.Baglio SR, Lagerweij T, Pérez-Lanzón M, et al. Blocking Tumor-Educated MSC Paracrine Activity Halts Osteosarcoma Progression. Clin Cancer Res. 2017;23(14):3721–3733. doi: 10.1158/1078-0432.CCR-16-2726. [DOI] [PubMed] [Google Scholar]
- 17.Roberts RD, Gross AC, Bid HK, Phelps D, Wedekind MF, Houghton PJ. Autocrine and paracrine IL-6 and IL-8 drive osteosarcoma lung tropism and facilitate metastasis [Abstract]. Cancer Res. 2016;76(5 Supplement):B40–B40. doi: 10.1158/1538-7445.PEDCA15-B40. [DOI] [Google Scholar]
- 18.Gross AC, Cam H, Phelps DA, et al. IL-6 and CXCL8 mediate osteosarcoma-lung interactions critical to metastasis. JCI Insight. 2018;3(16). doi: 10.1172/JCI.INSIGHT.99791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lizardo MM, Morrow JJ, Miller TE, et al. Upregulation of Glucose-Regulated Protein 78 in Metastatic Cancer Cells Is Necessary for Lung Metastasis Progression. Neoplasia. 2016;18(11):699–710. doi: 10.1016/j.neo.2016.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Jiang C, Chen H, Shao L, Dong Y. GRM4 gene polymorphism is associated with susceptibility and prognosis of osteosarcoma in a Chinese Han population. Med Oncol. 2014;31(7):50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Savage SA, Mirabello L, Wang Z, et al. Genome-wide association study identifies two susceptibility loci for osteosarcoma. Nat Genet. 2013;45(7):799–803. doi: 10.1038/ng.2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yang Y, Basu S, Mirabello L, Spector L, Zhang L. A Bayesian Gene-Based Genome-Wide Association Study Analysis of Osteosarcoma Trio Data Using a Hierarchically Structured Prior. Cancer Inform. 2018;17:1176935118775103. doi: 10.1177/1176935118775103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Behjati S, Tarpey PS, Haase K, et al. Recurrent mutation of IGF signalling genes and distinct patterns of genomic rearrangement in osteosarcoma. Nat Commun. 2017;8:15936. doi: 10.1038/ncomms15936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gröbner SN, Worst BC, Weischenfeldt J, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555(7696):321–327. doi: 10.1038/nature25480. [DOI] [PubMed] [Google Scholar]
- 25.Morrow JJ, Bayles I, Funnell APW, et al. Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat Med. 2018;24(2):176–185. doi: 10.1038/nm.4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pourebrahim R, Zhang Y, Liu B, et al. Integrative genome analysis of somatic p53 mutant osteosarcomas identifies Ets2-dependent regulation of small nucleolar RNAs by mutant p53 protein. Genes Dev. 2017;31(18):1847–1857. doi: 10.1101/gad.304972.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Brabetz S, Leary SES, Gröbner SN, et al. A biobank of patient-derived pediatric brain tumor models. Nat Med. 2018;24(11):1752–1761. doi: 10.1038/s41591-018-0207-3. [DOI] [PubMed] [Google Scholar]
- 28.Wang LL, Levy ML, Lewis RA, et al. Clinical manifestations in a cohort of 41 Rothmund-Thomson syndrome patients. Am J Med Genet. 2001;102(1):11–17. [DOI] [PubMed] [Google Scholar]
- 29.Wang LL, Levy ML, Lewis RA, et al. Clinical manifestations in a cohort of 41 Rothmund-Thomson syndrome patients. Am J Med Genet. 2001;102(1):11–17. [DOI] [PubMed] [Google Scholar]
- 30.Gianferante DM, Mirabello L, Savage SA. Germline and somatic genetics of osteosarcoma - connecting aetiology, biology and therapy. Nat Rev Endocrinol. 2017;13(8):480–491. doi: 10.1038/nrendo.2017.16. [DOI] [PubMed] [Google Scholar]
- 31.McBride KA, Ballinger ML, Killick E, et al. Li-Fraumeni syndrome: cancer risk assessment and clinical management. Nat Rev Clin Oncol. 2014;11(5):260–271. doi: 10.1038/nrclinonc.2014.41. [DOI] [PubMed] [Google Scholar]
- 32.Mai PL, Best AF, Peters JA, et al. Risks of first and subsequent cancers among TP53 mutation carriers in the National Cancer Institute Li-Fraumeni syndrome cohort. Cancer. 2016;122(23):3673–3681. doi: 10.1002/cncr.30248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gianferante DM, Mirabello L, Savage SA. Germline and somatic genetics of osteosarcoma - connecting aetiology, biology and therapy. Nat Rev Endocrinol. 2017;13(8):480–491. doi: 10.1038/nrendo.2017.16. [DOI] [PubMed] [Google Scholar]
- 34.Mirabello L, Yeager M, Mai PL, et al. Germline TP53 variants and susceptibility to osteosarcoma. J Natl Cancer Inst. 2015;107(7). doi: 10.1093/jnci/djv101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hettmer S, Archer NM, Somers GR, et al. Anaplastic rhabdomyosarcoma in TP53 germline mutation carriers. Cancer. 2014;120(7):1068–1075. doi: 10.1002/cncr.28507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhukova N, Ramaswamy V, Remke M, et al. Subgroup-specific prognostic implications of TP53 mutation in medulloblastoma. J Clin Oncol. 2013;31(23):2927–2935. doi: 10.1200/JCO.2012.48.5052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bougeard G, Renaux-Petel M, Flaman J-M, et al. Revisiting Li-Fraumeni Syndrome From TP53 Mutation Carriers. J Clin Oncol. 2015;33(21):2345–2352. doi: 10.1200/JCO.2014.59.5728. [DOI] [PubMed] [Google Scholar]
- 38.Zhao S, Kurenbekova L, Gao Y, et al. NKD2, a negative regulator of Wnt signaling, suppresses tumor growth and metastasis in osteosarcoma. Oncogene. 2015;34(39):5069–5079. doi: 10.1038/onc.2014.429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lauvrak SU, Munthe E, Kresse SH, et al. Functional characterisation of osteosarcoma cell lines and identification of mRNAs and miRNAs associated with aggressive cancer phenotypes. Br J Cancer. 2013;109(8):2228–2236. doi: 10.1038/bjc.2013.549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Neale G, Su X, Morton CL, et al. Molecular characterization of the pediatric preclinical testing panel. Clin Cancer Res. 2008;14(14):4572–4583. doi: 10.1158/1078-0432.CCR-07-5090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Stewart E, Federico S, Karlstrom A, et al. The Childhood Solid Tumor Network: A new resource for the developmental biology and oncology research communities. Dev Biol. 2016;411(2):287–293. doi: 10.1016/j.ydbio.2015.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Stewart E, Federico SM, Chen X, et al. Orthotopic patient-derived xenografts of paediatric solid tumours. Nature. 2017;549(7670):96–100. doi: 10.1038/nature23647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Guijarro MV, Ghivizzani SC, Gibbs CP. Animal models in osteosarcoma. Front Oncol. 2014;4:189. doi: 10.3389/fonc.2014.00189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Jones KB. Osteosarcomagenesis: modeling cancer initiation in the mouse. Sarcoma. 2011;2011:694136. doi: 10.1155/2011/694136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Gupte A, Baker EK, Wan S-S, et al. Systematic Screening Identifies Dual PI3K and mTOR Inhibition as a Conserved Therapeutic Vulnerability in Osteosarcoma. Clin Cancer Res. 2015;21(14):3216–3229. doi: 10.1158/1078-0432.CCR-14-3026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Paoloni M, Davis S, Lana S, et al. Canine tumor cross-species genomics uncovers targets linked to osteosarcoma progression. BMC Genomics. 2009;10(1):625. doi: 10.1186/1471-2164-10-625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yang Y, Yang R, Roth M, et al. Genetically transforming human osteoblasts to sarcoma: development of an osteosarcoma model. Genes Cancer. 2017;8(1–2):484–494. doi: 10.18632/genesandcancer.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fenger JM, London CA, Kisseberth WC. Canine osteosarcoma: a naturally occurring disease to inform pediatric oncology. ILAR J. 2014;55(1):69–85. doi: 10.1093/ilar/ilu009. [DOI] [PubMed] [Google Scholar]
- 49.Angstadt AY, Thayanithy V, Subramanian S, Modiano JF, Breen M. A genome-wide approach to comparative oncology: high-resolution oligonucleotide aCGH of canine and human osteosarcoma pinpoints shared microaberrations. Cancer Genet. 2012;205(11):572–587. doi: 10.1016/j.cancergen.2012.09.005. [DOI] [PubMed] [Google Scholar]
- 50.Kurzman ID, MacEwen EG, Rosenthal RC, et al. Adjuvant therapy for osteosarcoma in dogs: results of randomized clinical trials using combined liposome-encapsulated muramyl tripeptide and cisplatin. Clin Cancer Res. 1995;1(12):1595–1601. [PubMed] [Google Scholar]
- 51.Mason NJ, Gnanandarajah JS, Engiles JB, et al. Immunotherapy with a HER2-Targeting Listeria Induces HER2-Specific Immunity and Demonstrates Potential Therapeutic Effects in a Phase I Trial in Canine Osteosarcoma. Clin Cancer Res. 2016;22(17):4380–4390. doi: 10.1158/1078-0432.CCR-16-0088. [DOI] [PubMed] [Google Scholar]
- 52.Fan TM, Khanna C. Comparative Aspects of Osteosarcoma Pathogenesis in Humans and Dogs. Vet Sci. 2015;2(3):210–230. doi: 10.3390/vetsci2030210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Khanna C, Fan TM, Gorlick R, et al. Towards a Drug Development Path that Targets Metastatic Progression in Osteosarcoma. Clin Cancer Res. May 2014. doi: 10.1158/1078-0432.CCR-13-2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Aljubran AH, Griffin A, Pintilie M, Blackstein M. Osteosarcoma in adolescents and adults: survival analysis with and without lung metastases. Ann Oncol. 2009;20(6):1136–1141. doi: 10.1093/annonc/mdn731. [DOI] [PubMed] [Google Scholar]
- 55.Qiu H, Orr FW, Jensen D, et al. Arrest of B16 melanoma cells in the mouse pulmonary microcirculation induces endothelial nitric oxide synthase-dependent nitric oxide release that is cytotoxic to the tumor cells. Am J Pathol. 2003;162(2):403–412. doi: 10.1016/S0002-9440(10)63835-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Piskounova E, Agathocleous M, Murphy MM, et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature. 2015;527(7577):186–191. doi: 10.1038/nature15726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fidler IJ. Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125 I-5-iodo-2’-deoxyuridine. J Natl Cancer Inst. 1970;45(4):773–782. [PubMed] [Google Scholar]
- 58.Cameron MD, Schmidt EE, Kerkvliet N, et al. Temporal progression of metastasis in lung: cell survival, dormancy, and location dependence of metastatic inefficiency. Cancer Res. 2000;60(9):2541–2546. [PubMed] [Google Scholar]
- 59.Weiss L, Mayhew E, Rapp DG, Holmes JC. Metastatic inefficiency in mice bearing B16 melanomas. Br J Cancer. 1982;45(1):44–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ren L, Mendoza A, Zhu J, et al. Characterization of the metastatic phenotype of a panel of established osteosarcoma cells. Oncotarget. 2015;6(30):29469–29481. doi: 10.18632/oncotarget.5177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Koshkina NV, Khanna C, Mendoza A, Guan H, DeLauter L, Kleinerman ES. Fas-negative osteosarcoma tumor cells are selected during metastasis to the lungs: the role of the Fas pathway in the metastatic process of osteosarcoma. Mol Cancer Res. 2007;5(10):991–999. doi: 10.1158/1541-7786.MCR-07-0007. [DOI] [PubMed] [Google Scholar]
- 62.Khanna C, Wan X, Bose S, et al. The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis. Nat Med. 2004;10(2):182–186. doi: 10.1038/nm982. [DOI] [PubMed] [Google Scholar]
- 63.Tsai YC, Mendoza A, Mariano JM, et al. The ubiquitin ligase gp78 promotes sarcoma metastasis by targeting KAI1 for degradation. Nat Med. 2007;13(12):1504–1509. doi: 10.1038/nm1686. [DOI] [PubMed] [Google Scholar]
- 64.Bid HK, Roberts RD, Cam M, et al. ΔNp63 Promotes Pediatric Neuroblastoma and Osteosarcoma by Regulating Tumor Angiogenesis. Cancer Res. 2014;74(1):320–329. doi: 10.1158/0008-5472.CAN-13-0894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shimizu T, Sugihara E, Yamaguchi-Iwai S, et al. IGF2 preserves osteosarcoma cell survival by creating an autophagic state of dormancy that protects cells against chemotherapeutic stress. Cancer Res. 2014;74(22):6531–6541. doi: 10.1158/0008-5472.CAN-14-0914. [DOI] [PubMed] [Google Scholar]
- 66.Almog N, Ma L, Raychowdhury R, et al. Transcriptional switch of dormant tumors to fast-growing angiogenic phenotype. Cancer Res. 2009;69(3):836–844. doi: 10.1158/0008-5472.CAN-08-2590. [DOI] [PubMed] [Google Scholar]
- 67.Tiram G, Segal E, Krivitsky A, et al. Identification of Dormancy-Associated MicroRNAs for the Design of Osteosarcoma-Targeted Dendritic Polyglycerol Nanopolyplexes. ACS Nano. 2016;10(2):2028–2045. doi: 10.1021/acsnano.5b06189. [DOI] [PubMed] [Google Scholar]
- 68.Mendoza A, Hong S-H, Osborne T, et al. Modeling metastasis biology and therapy in real time in the mouse lung. J Clin Invest. 2010;120(8):2979–2988. doi: 10.1172/JCI40252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Young ED, Strom K, Tsue AF, et al. Automated quantitative image analysis for ex vivo metastasis assays reveals differing lung composition requirements for metastasis suppression by KISS1. Clin Exp Metastasis. 2018;35(1–2):77–86. doi: 10.1007/s10585-018-9882-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lizardo MM, Sorensen PH. Practical Considerations in Studying Metastatic Lung Colonization in Osteosarcoma Using the Pulmonary Metastasis Assay. J Vis Exp. 2018;(133). doi: 10.3791/56332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Entenberg D, Voiculescu S, Guo P, et al. A permanent window for the murine lung enables high-resolution imaging of cancer metastasis. Nat Methods. 2018;15(1):73–80. doi: 10.1038/nmeth.4511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Bouffet E, Larouche V, Campbell BB, et al. Immune Checkpoint Inhibition for Hypermutant Glioblastoma Multiforme Resulting From Germline Biallelic Mismatch Repair Deficiency. J Clin Oncol. 2016;34(19):2206–2211. doi: 10.1200/JCO.2016.66.6552. [DOI] [PubMed] [Google Scholar]
- 73.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–413. doi: 10.1126/science.aan6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hellmann MD, Callahan MK, Awad MM, et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell. 2018;33(5):853–861.e4. doi: 10.1016/j.ccell.2018.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.McCarthy EF. The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop J. 2006;26:154–158. [PMC free article] [PubMed] [Google Scholar]
- 76.Kovac M, Blattmann C, Ribi S, et al. Exome sequencing of osteosarcoma reveals mutation signatures reminiscent of BRCA deficiency. Nat Commun. 2015;6(1):8940. doi: 10.1038/ncomms9940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Man T-K, Lu X-Y, Jaeweon K, et al. Genome-wide array comparative genomic hybridization analysis reveals distinct amplifications in osteosarcoma. BMC Cancer. 2004;4(1):45. doi: 10.1186/1471-2407-4-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Koirala P, Roth ME, Gill J, et al. Immune infiltration and PD-L1 expression in the tumor microenvironment are prognostic in osteosarcoma. Sci Rep. 2016;6(1):30093. doi: 10.1038/srep30093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hingorani P, Maas ML, Gustafson MP, et al. Increased CTLA-4(+) T cells and an increased ratio of monocytes with loss of class II (CD14(+) HLA-DR(lo/neg)) found in aggressive pediatric sarcoma patients. J Immunother cancer. 2015;3(1):35. doi: 10.1186/s40425-015-0082-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Heymann M-F, Lézot F, Heymann D. The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma. Cell Immunol. November 2017. doi: 10.1016/j.cellimm.2017.10.011. [DOI] [PubMed] [Google Scholar]
- 81.Lussier DM, O’Neill L, Nieves LM, et al. Enhanced T-cell immunity to osteosarcoma through antibody blockade of PD-1/PD-L1 interactions. J Immunother. 2015;38(3):96–106. doi: 10.1097/CJI.0000000000000065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lussier DM, Johnson JL, Hingorani P, Blattman JN. Combination immunotherapy with α-CTLA-4 and α-PD-L1 antibody blockade prevents immune escape and leads to complete control of metastatic osteosarcoma. J Immunother Cancer. 2015;3(1):21. doi: 10.1186/s40425-015-0067-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Takahashi Y, Yasui T, Tamari K, et al. Radiation enhanced the local and distant anti-tumor efficacy in dual immune checkpoint blockade therapy in osteosarcoma. PLoS One. 2017;12(12):e0189697. doi: 10.1371/journal.pone.0189697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ratti C, Botti L, Cancila V, et al. Trabectedin Overrides Osteosarcoma Differentiative Block and Reprograms the Tumor Immune Environment Enabling Effective Combination with Immune Checkpoint Inhibitors. Clin Cancer Res. 2017;23(17):5149–5161. doi: 10.1158/1078-0432.CCR-16-3186. [DOI] [PubMed] [Google Scholar]
- 85.Tawbi HA, Burgess M, Bolejack V, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017;18(11):1493–1501. doi: 10.1016/S1470-2045(17)30624-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gonzalez L, Strbo N, Podack ER. Humanized mice: novel model for studying mechanisms of human immune-based therapies. Immunol Res. 2013;57(1–3):326–334. doi: 10.1007/s12026-013-8471-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Whelan JS, Bielack SS, Marina N, et al. EURAMOS-1, an international randomised study for osteosarcoma: results from pre-randomisation treatment. Ann Oncol Off J Eur Soc Med Oncol. 2015;26(2):407–414. doi: 10.1093/annonc/mdu526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Wendler D, Jenkins T. Children’s and Their Parents’ Views on Facing Research Risks for the Benefit of Others. Arch Pediatr Adolesc Med. 2008;162(1):9. doi: 10.1001/archpediatrics.2007.3. [DOI] [PubMed] [Google Scholar]
- 89.Berg SL, Winick N, Ingle AM, Adamson PC, Blaney SM. Reasons for participation in optional pharmacokinetic studies in children with cancer: A Children’s Oncology Group phase 1 consortium study. Pediatr Blood Cancer. 2010;55(1):n/a–n/a. doi: 10.1002/pbc.22529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lagmay JP, Krailo MD, Dang H, et al. Outcome of Patients With Recurrent Osteosarcoma Enrolled in Seven Phase II Trials Through Children’s Cancer Group, Pediatric Oncology Group, and Children’s Oncology Group: Learning From the Past to Move Forward. J Clin Oncol. 2016;34(25):3031–3038. doi: 10.1200/JCO.2015.65.5381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kleinerman E Maximum benefit of chemotherapy for osteosarcoma achieved-what are the next steps? Lancet Oncol. 2016;17(10):1340–1342. doi: 10.1016/S1470-2045(16)30270-4. [DOI] [PubMed] [Google Scholar]
- 92.Guenther LM, Rowe RG, Acharya PT, et al. Response Evaluation Criteria in Solid Tumors (RECIST) following neoadjuvant chemotherapy in osteosarcoma. Pediatr Blood Cancer. 2018;65(4):e26896. doi: 10.1002/pbc.26896. [DOI] [PubMed] [Google Scholar]
- 93.Isakoff MS, Goldsby R, Villaluna D, et al. A phase II study of eribulin in recurrent or refractory osteosarcoma: A report from the Children’s Oncology Group. Pediatr Blood Cancer. 2019;66(2):e27524. doi: 10.1002/pbc.27524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Isakoff MS, Goldsby R, Villaluna D, et al. Rapid Protocol Enrollment in Osteosarcoma: A Report From the Children’s Oncology Group. Pediatr Blood Cancer. 2016;63(2):370–371. doi: 10.1002/pbc.25754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Hampson LV, Whitehead J, Eleftheriou D, Brogan P. Bayesian methods for the design and interpretation of clinical trials in very rare diseases. Stat Med. 2014;33(24):4186–4201. doi: 10.1002/sim.6225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Murphy B, Yin H, Maris JM, et al. Evaluation of Alternative In Vivo Drug Screening Methodology: A Single Mouse Analysis. Cancer Res. 2016;76(19):5798–5809. doi: 10.1158/0008-5472.CAN-16-0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Fan TM, Khanna C. Comparative Aspects of Osteosarcoma Pathogenesis in Humans and Dogs. Vet Sci. 2015;2(3):210–230. doi: 10.3390/vetsci2030210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hingorani P, Janeway K, Crompton BD, et al. Current state of pediatric sarcoma biology and opportunities for future discovery: A report from the sarcoma translational research workshop. Cancer Genet. 2016;209(5):182–194. doi: 10.1016/j.cancergen.2016.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]