Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

REVIEwS

Advancing therapy for osteosarcoma


Jonathan Gill    and Richard Gorlick    ✉
Abstract | Improving the survival of patients with osteosarcoma has long proved challenging,
although the treatment of this disease is on the precipice of advancement. The increasing
feasibility of molecular profiling together with the creation of both robust model systems and
large, well-​annotated tissue banks has led to an increased understanding of osteosarcoma biology.
The historical invariability of survival outcomes and the limited number of agents known to be
active in the treatment of this disease facilitate clinical trials designed to identify efficacious novel
therapies using small cohorts of patients. In addition, trial designs will increasingly consider the
genetic background of the tumour through biomarker-​based patient selection, thereby enriching
for clinical activity. Indeed, osteosarcoma cells are known to express a number of surface proteins
that might be of therapeutic relevance, including B7-​H3, GD2 and HER2, which can be targeted
using antibody–drug conjugates and/or adoptive cell therapies. In addition, immune-​checkpoint
inhibition might augment the latter approach by helping to overcome the immunosuppressive
tumour microenvironment. In this Review, we provide a brief overview of current osteosarcoma
therapy before focusing on the biological insights from the molecular profiling and preclinical
modelling studies that have opened new therapeutic opportunities in this disease.

Despite being the most common primary bone cancer in presenting with localized disease4; however, limited
children and young adults, osteosarcoma is a very rare therapeutic progress has been made since that time. An
cancer, with approximately 400 new cases diagnosed increased understanding of the biology of the disease
annually in children and young adults in the USA1,2. has highlighted its heterogeneity and revealed molecu-
The incidence of the disease peaks in adolescence lar aberrations that define potential patient subgroups.
(4.4 cases per million individuals)1,2, which coincides These advances were made possible by the creation of
with the pubertal growth spurt. Osteosarcoma most well-​annotated tissue banks beginning at the turn
commonly arises in the metaphysis of the long bones of the century in parallel with the broader availability of
near the growth plates, with two-​thirds of tumours technologies for comprehensive molecular profiling.
emerging around the knee joint in the distal femur, fol- Many of the most common genetic alterations
lowed by the proximal tibia as the second most frequent detected in osteosarcomas, such as TP53 break-​apart
site; the proximal humerus is the third most common translocations and RB1 deletions that abrogate the
site, accounting for 10% of tumours3. More than 85% of expression of key tumour-​suppressor proteins, have
patients present with localized disease. Of the patients been challenging to target therapeutically. Nevertheless,
who present with metastatic disease, 74% present with small subsets of these tumours harbour potentially
lung-​only metastases, 9% with bone-​only metastases, targetable somatic alterations. More broadly, clinical
and 8% with both bone and lung metastases3. A second trial designs incorporating biomarker-​based selection
peak in the incidence of osteosarcoma occurs in adults might increase the proportion of patients with oste-
aged >65 years (4.2 cases per million individuals), often osarcoma who benefit from a particular treatment.
manifesting as secondary cancers (in 25% of patients) or Cellular differentiation within the mesenchymal cell
as a consequence of Paget disease (in 10%)1. This Review lineage is a characteristic feature of osteosarcoma and
is focused primarily on the treatment of osteosarcoma in results in the consistent expression of several cell-​surface
adolescent and young adult patients. The rarity of this molecules. Accordingly, novel antibody-​b ased and
Division of Pediatrics, cancer has necessitated collaborative efforts involving adoptive cell therapy approaches are broadening the
The University of Texas MD cooperative groups, such as the Children’s Oncology range of possible therapeutic targets in osteosarcoma;
Anderson Cancer Center,
Houston, TX, USA.
Group (COG) in the USA, as well as international immune-​checkpoint inhibitors (ICIs), although disap-
✉e-​mail: rgorlick@ collaborations to complete clinical trials. pointing as single agents in this disease, might have roles
mdanderson.org The standard therapy for osteosarcoma, comprising in enhancing the efficacy of such approaches.
https://doi.org/10.1038/ surgery and chemotherapy, was established in the 1980s Notably, robust preclinical models of osteosarcoma
s41571-021-00519-8 and results in long-​term survival in >60% of patients are now available and could potentially facilitate drug

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

Key points creates opportunities to rapidly evaluate novel thera-


pies in single-​arm clinical trials using small numbers
• Osteosarcoma is the most common primary malignant tumour of bone, with a peak of patients. The outcomes of patients with resectable
incidence in adolescents and young adults coinciding with the pubertal growth spurt. recurrent disease (typically pulmonary metastases) are
• Limited progress has been made in improving the survival outcomes in patients with also highly conducive to single-​arm study designs, with
osteosarcoma over the past four decades. a median EFS of 4 months and a 2-​year EFS of 12%17.
• Improved molecular characterization has revealed subcategories of osteosarcoma
that might enable a precision medicine approach with agents targeting key Genomic alterations and heterogeneity
alterations in a particular pathway. Since the advent of karyotyping, osteosarcomas have
• Tumour-​suppressor genes are commonly altered in this disease, particularly TP53 been known to be genomically complex18,19. Even before
(>90%) and RB1 (30%). Molecular targets include receptor tyrosine kinases, CDK4/6, analyses of chromosomes were widely performed, rou-
Aurora kinase B and DNA damage response pathways.
tine histological staining revealed substantial variation in
• Immune-​based targeted therapies, including monoclonal antibodies, antibody–drug the appearance of individual tumours, with the pattern
conjugates and chimeric antigen receptor T cells targeting cell-​surface proteins
of cellular differentiation defining the disease subtype20.
commonly overexpressed in osteosarcoma, are in active clinical development.
Conventionally, osteosarcomas are classified as osteo-
• Owing to advances in biological understanding, the development of robust
blastic (50%), chondroblastic (25%) or fibroblastic (25%)
preclinical models, the feasibility of rapid clinical testing and novel treatment
concepts, long-​awaited improvements in the outcomes of patients with
according to their predominant differentiation pattern.
osteosarcoma are anticipated in the near future. These classifications are somewhat arbitrary given that
the majority of tumours contain more than one histo-
logical pattern, which suggests considerable intra-​patient
development. Moreover, the standardization of trial heterogeneity21. The frequent occurrence of osteosar-
designs and efficacy end points has ensured that mul- coma in patients with cancer predisposition syndromes,
tiple groups are capable of conducting clinical studies such as Li–Fraumeni syndrome and hereditary retino-
rapidly and efficiently. These advances suggest the field blastoma, led to the early recognition of prevalent alter-
is on the verge of progress in the treatment of osteosar- ations in tumour-​suppressor genes in this malignancy,
coma. Hence, advances in osteosarcoma therapy serve including TP53 (>90%) and RB1 (30%)22. The identifi-
as the organizing principle of this Review, encompassing cation of these and other recurrently altered genes raised
current therapy, targeted treatments based on specific hopes that certain molecular alterations crucial to the
molecular alterations and immune-​based approaches. pathogenesis or drug sensitivity of osteosarcoma would
serve as prognostic or predictive biomarkers. In numer-
Current osteosarcoma therapy ous paediatric malignancies, including neuroblastoma
The established three-​drug chemotherapy regimens and acute lymphoblastic leukaemia, patients receive
used in the treatment of localized osteosarcoma has risk-​stratified treatment, often aided by the identification
led to consistent patient outcomes over the past four of prognostic molecular characteristics, such as MYC
decades. In North America, the most common chemo- amplification or the Philadelphia chromosome translo-
therapy regimen, MAP, consists of high-​dose methotrex- cation, respectively23,24. With this goal in mind, numer-
ate, doxorubicin (adriamycin) and cisplatin (platinol) ous institutional osteosarcoma tissue banks began to be
administered both before and after definitive surgical created and, in 1998, a national biobank was established
resection. In the field of osteosarcoma, the terminology in the USA by the COG. The COG Tissue Bank has been
of ‘neoadjuvant’ and ‘adjuvant’ chemotherapy has been described in several publications25,26, which highlight the
replaced with ‘induction’ and ‘consolidation’ treatment, broad availability of osteosarcoma samples as well as
respectively. Numerous clinical trials have tested vari- the philanthropic support of advocacy groups such as the
ous combinations of the five chemotherapeutic agents QuadW Foundation, which was a major contributor to
known to be active in this disease (methotrexate, doxo- the success of the initiative. Numerous other organiza-
rubicin, cisplatin, ifosfamide and etoposide), some using tions around the world have initiated biobanks including
pathological response at the time of definitive surgery to osteosarcoma, such as the Bone Cancer Research Trust
select consolidation therapy, without additional progress in the UK and EuroBoNeT in the European Union. With
being made5–9. These studies have revealed that frontline the continuous evolution of tissue banking, efforts do not
agents can be replaced with other active drugs but that have to be centred in a single institution. For example, the
adding more agents into the initial treatment regimen Count Me In initiative led by the Broad Institute of MIT
does not improve outcomes (Fig. 1) nor does adjusting and Harvard (with institutional review board approval
therapy based on pathological response. as an entity) aims to obtain tissue samples and data from
In patients with relapsed and/or metastatic osteosar- patients directly and includes a specific Osteosarcoma
coma, surgery with metastasectomy has been shown to Project component. These tissue banks have facilitated a
provide survival benefit10–12. Chemotherapy regimens large number of molecular profiling studies of osteosar-
comprising ifosfamide and etoposide or gemcitabine coma, predominantly focused on limited sets of genes27,
and docetaxel have some efficacy in patients with unre- although, unfortunately, a reproducible prognostic factor
sectable relapsed disease13–15. Data from several phase II has not been identified thus far.
trials of various treatments demonstrate the dismal Patterns in data emerging from comprehensive
outcomes of patients with relapsed, unresectable oste- molecular profiling studies have prompted a paradigm
osarcoma, with an aggregate event-​free survival (EFS) shift towards broader analyses of pathways and genomic
of only 12% at 4 months16; however, this situation signatures, rather than of individual genes, to identify

www.nature.com/nrclinonc

0123456789();:
Reviews

100
Mean EFS with
four-drug regimens157
COSS-86 (MAPI)158 90
IOR/OS-4 (MAPI)159
POG-8651 (MAP BCD)160
INT-0133 (MAPI)161 80

ISG-SSG-1 (MAPI) 162

EOI-80861 (AP V BCD)163


70

Mean EFS with


three-drug regimens157
60
INT-0133 (MAP)161
MIOS (MAP)164
EFS (%)

MEI165 50
API8
COSS-80 (MAP)166
40
COSS-82 (MAP)167
EOI-80831 (MAP)168
SFOP-OS94 (MEI)169 30
IOR/OS-2 (MAP)170
IOR/OS-3 (MAP)170
SSG-VIII (MAP)171 20

Mean EFS with


two-drug regimens157 10
EOI-80861 (AP)163
EOI-80931 (AP)163
0
SFOP-OS94 (MA)169 0 1 2 3 4 5
Follow-up duration (years)

Fig. 1 | EFS with chemotherapy regimens comprising two, three or four drugs in patients with osteosarcoma.
The dashed lines depict the event-​free survival (EFS) curves from numerous large-​cohort trials of various chemotherapy
regimens in patients with osteosarcoma. The aggregate EFS curves of trials investigating combinations of two, three
or four cytotoxic agents are shown using red, blue and green bold lines, respectively. Notably, the green EFS curves
corresponding to four-​drug regimens overlap substantially with the blue lines relating to three-​drug regimens. Thus,
the outcomes of patients treated with various three-​drug or four-​drug regimens are effectively the same; however,
patients treated with two-​drug combinations have inferior outcomes (red lines). A, doxorubicin; B, bleomycin;
C, cyclophosphamide; D, dactinomycin; E, etoposide; I, ifosfamide; M, methotrexate; P, cisplatin; V, vincristine157–171.

pathogenetic aberrations28–33. In a seminal publication a few shared structural variations22. However, certain
describing chromothripsis34, whereby chromosomal genes were commonly altered, albeit through different
shattering results in numerous simultaneous genomic somatic alterations, including the tumour-​suppressor
rearrangements, osteosarcoma was identified as one of genes TP53 (95%), RB1 (29%), ATRX (52%) and
the tumour types in which this phenomenon occurs. DLG2 (52%)22. In addition to the structural alterations
Indeed, chromothripsis is now recognized as a com- observed, kataegis was observed in 50% of the samples22.
mon oncogenic event in osteosarcomas — detectable A whole-​exome sequencing (WES) study revealed alter-
in up to 75% of tumours — and can involve multiple ations in the same genes, although the unique mutations
chromosomes34,35. The loss of an organized chromosome identified in each sample underscore the substantial
structure has complicated the identification of genetic interpatient heterogeneity of this disease29. In a separate
commonalities across osteosarcomas, with multiple pub- WES study30, a genomic instability signature character-
lished sequencing studies revealing similar findings but istic of BRCA1/2-deficient tumours was observed in 80%
focusing on different perturbed pathways or genomic of osteosarcomas.
signatures. For example, some groups have focused on With regard to specific signalling pathways, altera-
altered tumour-​suppressor genes22,29,30,36, some on par- tions in IGF1, IGF1R, IGF2R, IGFBP5 or components of
ticular signalling pathways31,32 and others on the immune the downstream signalling pathway have been detected
landscape of the disease28,33,37. in 27% of 112 osteosarcomas through WGS or WES,
In a whole-​genome sequencing (WGS) study, differ- with high-​level IGF1R amplifications (≥15 copies)
ences in the number and location of structural altera- detected by fluorescence in situ hybridization in 14% of
tions were observed across most osteosarcomas, with an additional 87 samples31. In another study32, pathway

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

analyses and other computational or experimental found to have multiple pathway alterations that could be
approaches were used to decipher the heterogeneity categorized into more than one group, whereas others
of aberrations observed in complementary WGS, WES were not easily classified into any of the groups (R.G.,
and RNA sequencing data from patients with osteosar- unpublished data) (Fig. 2). On the whole, the genomic
coma. The results indicate the convergence of unique complexity and heterogeneity observed in multiple
alterations on the activation of the PI3K–ATK–mTOR studies indicate that osteosarcoma might need to be
pathway32. Combined genomic and transcriptomic considered as more than one disease.
analyses have also enabled the identification of multiple
fusion transcripts that only rarely correlate directly with Models of osteosarcoma
chromosomal rearrangements in osteosarcoma sam- Over the past 15 years, >100 drugs have been screened for
ples, suggesting the involvement of post-​transcription activity in osteosarcoma using PDX models developed
trans-​splicing36. These fusion transcripts, although by the PPTC41 (Figs 2,3; Supplementary Table 1). More
involving multiple loci in different cell lines and recently, a European consortium has established the
patient samples, most commonly affect the TP53 locus Innovative Therapies for Children with Cancer Paediatric
and serve as another mechanism by which TP53 is Preclinical Proof-​of-​c oncept Platform (ITCC P4),
inactivated in osteosarcoma36. which is seeking to establish and fully characterize 400
A number of studies have evaluated the immuno­ new PDX models of high-​risk paediatric solid tumours,
genomic landscape of osteosarcoma. The resulting data including osteosarcomas. Indeed, in the aforemen-
demonstrate that osteosarcomas can have an immune tioned study by the Sweet-​Cordero group39, common
‘hot’ or ‘cold’ signature or an intermediate level of osteosarcoma-​associated molecular alterations were
immune cell infiltration33. Most of the cold tumours accurately recapitulated in a set of PDX models and
demonstrated a substantial number of genes with copy responses to genome-​informed treatments were seen in
number losses and decreased expression of HLA genes, all PDXs tested. The PPTC have also sequenced and char-
leading to immune escape37. The hot subset demonstrated acterized 31 PDX models of osteosarcoma, demonstrat-
an adaptive immune resistance mechanism involving ing substantial tumour heterogeneity that recapitulates
upregulation of T cell inhibitory immune-​checkpoint the high prevalence of complex genomic rearrangements
proteins, including PD-​L1 and CTLA4 (ref.37). Notably, described in patients with osteosarcoma42.
publicly available deep-​sequencing data from 88 patients Despite sharing the same genomic alterations as
with osteosarcoma, which were generated as part of the their human tumour counterparts, PDX models have
US National Cancer Institute (NCI) Therapeutically clear limitations. In particular, therapeutic activity in
Applicable Research to Generate Effective Treatments these models does not necessarily translate into efficacy
(TARGET) Osteosarcoma project, have been used to in clinical trials (Fig. 2). For example, both glembatu-
generate a signature comprising 13 immune-​related mumab vedotin (a transmembrane glycoprotein NMB
genes that are prognostic of distant metastasis and (GPNMB)-​targeted antibody–drug conjugate (ADC)
overall survival (OS)28. with a microtubule inhibitor payload) and the cyto-
In public presentations, TARGET Osteosarcoma pro- toxic agent eribulin (also a microtubule inhibitor) were
ject investigator Paul Meltzer has made the analogy that predicted to have activity against osteosarcoma based
the mutational profiles of osteosarcomas are compara- on PDX modelling (Fig. 3) but ultimately had limited
ble to snowflakes38: like snowflakes, all osteosarcomas efficacy in patients with relapsed osteosarcoma43,44. In
share a common recognizable ‘shape’ but each tumour is the case of eribulin, this disparity probably reflects a
unique. Taken together, however, these studies demon- failure to fully account for pharmacokinetic differences
strate how an explosion of knowledge is yielding an between mice and humans45. A major concern is that
understanding of common patterns visible through the PDX tumours are necessarily implanted in immunode-
enormous genomic complexity of osteosarcoma. ficient mice; therefore, these models fail to recapitulate
the immunological aspects of cancers and cancer treat-
Subcategorization of osteosarcomas ments, which in particular poses substantial challenges
In a landmark study, the Sweet-​C ordero group 39 to the testing of immunotherapies. Defining the level of
attempted to establish some organization in the inter- activity in PDX models that translates into efficacy in
patient heterogeneity of osteosarcoma by defining dis- clinical trials is also problematic: should this be deter-
ease subclassifications based on pathway alterations. mined by the proportion of models demonstrating a
Using deep-​sequencing to define genomic alterations, response or by the degree of response within a single
osteosarcoma could be subclassified into six groups model? This question is confounded by the fact that the
according to the key signalling pathways activated: lack of activity of any novel agents tested in patients to
cyclin E1/CDK2, MYC/CDK9, CDK4/CDK6/FOXM1, date limits the ability to extrapolate a positive predictive
PTEN/PI3K/AKT1/mTOR, AURKB and VEGFA/ value from the preclinical models.
VEGFR39 (Fig. 2). While conceptually deriving from In addition to PDX models, numerous transgenic
approaches taken in classifying other cancer types, such models of osteosarcoma have been generated but,
as medulloblastoma40, it must be acknowledged that notably, have rarely been used for drug testing. These
more overlap occurs in the context of osteosarcoma. In transgenic models include Tp53 and combined Tp53
an analysis of patient-​derived xenograft (PDX) models and Rb1 knockouts driven by an osteoblast-​based pro-
developed by the NCI Paediatric Preclinical Testing moter, which result in osteosarcomas in 77% and 100%
Consortium (PPTC), for example, some tumours were of mice, respectively46,47. One potential flaw of these

www.nature.com/nrclinonc

0123456789();:
Reviews

a e
MYC activation VEGF VEGF–VEGFR
pathway alterations
Predicted Response
responder observed Predicted Response
PIP2 PIP3 responder observed
OS1 c PI3K
P P
OS2 P P OS1
CDK4 or FOXM1
upregulation OS2
OS9 VEGFR
Cyclin D PTEN AKT OS9
OS17 Predicted Response
CDK4 responder observed OS17
OS31 d PTEN–PI3K–AKT
OS1 pathway alterations OS31
OS33
P OS2 Predicted Response OS33
MYC RB1 OS9 responder observed
OS17 OS1
OS31 OS2
OS33 OS9
OS17
FOXM1
Cyclin E1 OS31
f
CDK2 OS33
b Overexpression of
CCNE1 amplification AURKA or AURKB
Predicted Response Predicted Response
responder observed responder observed
OS1 OS1
Aurora A
OS2 OS2
OS9 Aurora B OS9
OS17 OS17
OS31 OS31
OS33 OS33

MCR CR PR SD PD2 PD1 Not tested

Fig. 2 | Molecular categories of osteosarcoma and responses to the no activity in these models. e | The one model predicted to respond to
corresponding molecularly targeted treatments in PPTC PDX models. agents targeting the VEGF–VEGFR signalling pathway (OS17) was indeed
The National Cancer Institute Paediatric Preclinical Testing Consortium responsive to VEGFR-​directed tyrosine kinase inhibitors. f | In addition, SD
(PPTC) patient-​derived xenograft (PDX) models of osteosarcoma (see was observed in a model (OS33) that was not expected to respond. Two
Supplementary Table 1) that are predicted to respond to specific targeted models (OS2 and OS33) were predicted to respond to the Aurora kinase
therapies based on their characteristic genomic alterations are indicated inhibitor MN8237 owing to the overexpression of AURKA or AURKB;
in green in the first column of each table. The actual responses observed in however, responses observed in the PPTC PDX models did not correlate
each of the PDX models are indicated in the second column of each table. with these genomic alterations. Note that some models (such as OS1 and
a | Two models (OS2 and OS17) have activation of MYC signalling and are OS17) carry genomic alterations that predict responses to various
therefore predicted to respond to agents targeting this pathway, although inhibitors of multiple pathways, suggesting that combination therapy
such agents have not yet been tested by the PPTC. b | Three models (OS1, might be warranted. CR, complete response (disappearance of measurable
OS9 and OS17) were predicted to respond to CDK2 inhibitors, owing to tumour mass for at least one time point); MCR, maintained complete
CCNE1 amplification; however, activity was observed only in the OS1 response (tumour volume <0.10 cm3 at the end of the study period); PD1,
model in the form of SD. c | Two models (OS1 and OS17) have upregulation progressive disease (>25% increase in tumour volume at the end of the
of CDK4 and/or FOXM1 and are therefore predicted to respond to CDK4/6 study period); PD2, delayed progressive disease (same as PD1 but with
inhibitors, which have not been tested by the PPTC to date. d | PTEN loss or tumour growth delay); PR, partial response (≥50% tumour regression for at
activating AKT1 mutations in three models (OS1, OS9 and OS17) were least one time point but with measurable tumour); SD, stable disease
predicted to confer sensitivity to the inhibition of the PI3K–AKT–mTOR (<50% regression and ≤25% increase in tumour volume at the end of the
signalling pathway, although the AKT inhibitor MK-2206 had minimal or study period).

models relates to their propensity for the development overexpressed in 41% of human osteosarcomas and this
of tumours in the jaw and head, which typically occur in feature is associated with an increased risk of metasta-
only 9% of patients with osteosarcoma46,47. A transgenic sis as well as with unfavourable survival outcomes49,50.
model in which the expression of Myc is driven by the Historically, Fos transgenes, as well as radiation and
lymphocyte-​specific enhancer EµSRα in the presence chronic parathyroid hormone administration, have also
of tetracycline leads to osteosarcomas in 1% of mice, been used to induce osteosarcoma formation in mice51,52.
presumably owing to the activation of the enhancer These various transgenic models enable the evaluation
element in immature osteoblasts; the resulting tumours of osteosarcomas in a native microenvironment, which
demonstrate oncogene addiction to Myc48. MYC is mitigates some of the issues associated with PDX models.

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

ab
m u
ec b

tin um
xt a
an

tin ma
el X

ru um

D
n

an

in

in
od D

ca

do at

PB

tin
do ta
ec

in

in
de stuz

i st

i st
m CP

ve mb
te

ve mro

6-

la
ul

ul
ot

cr

cr
po

sp
T

27
ib

ib
le
a

n
PP

Sa
Tr

To

Ci
Iri

Er

Er
Vi

Vi
G

m
OS1
OS2
OS9
OS17
OS31
OS33

MCR CR PR SD PD2 PD1 Not tested

Fig. 3 | Comparisons of the activity of aDCs and drugs similar to the deruxtecan and samrotamab vedotin), which might reflect resistance to the
aDC payloads in PDX models of osteosarcoma. In the National Cancer payload of the ADC. Conversely, some models might be less sensitive to
Institute Paediatric Preclinical Testing Consortium (PPTC) patient-​derived the ADC than the free drug owing to potential differences in the expression
xenograft (PDX) models of osteosarcoma (see Supplementary Table 1), of the target antigen. CR, complete response (disappearance of measurable
variable responses are observed between antibody–drug conjugates (ADCs) tumour mass for at least one time point); MCR, maintained complete
targeting HER2 (trastuzumab deruxtecan), LRRC15 (samrotamab vedotin; response (tumour volume <0.10 cm3 at the end of the study period); PD1,
ABBV-805), GPNMB (glembatumumab vedotin), or B7-​H3 (m276-​PBD) and progressive disease (>25% increase in tumour volume at the end of the
unconjugated drugs with comparable mechanisms of actions to the ADC study period); PD2, delayed progressive disease (same as PD1, but with
payloads. This disparity suggests that the activity of ADCs is at least partially tumour growth delay); PR, partial response (≥50% tumour regression for at
driven by the selective, antibody-​mediated targeting of the drug to least one time point, but with measurable tumour); SD, stable disease (<50%
tumours. Nevertheless, some models are resistant regardless of the ADC regression and ≤25% increase in tumour volume at the end of the study
target (for example, the OS1 model is resistant to both trastuzumab period).

However, transgenic models might still not accurately models is required to create a responder hypothesis to
recapitulate immune responses to osteosarcomas in enrich for pathway alterations present in specific models.
patients owing to the differences between the mouse and This process is confounded by the fact that some PDX
human immune systems. An alternative strategy to over- models have alterations in multiple pathways (Fig. 2).
come this potential issue involves the creation of PDX Understanding the effects of overlapping pathway alter-
models of osteosarcoma in humanized mice53. This field ations and responsiveness to targeted therapies will
is becoming more complex and a range of models with require testing with both single agents and combination
varying degrees of immune reconstitution are becoming treatments in order to identify driver versus bystander
available54. With humanized PDX models, the haemato- alterations. In addition, the complexity of the pathway
poietic stem cells used for immune reconstitution should alterations underscores the need to expand the number
ideally be derived from the same donor patient to avoid of PDX models being utilized in order to better encap-
graft-​versus-​tumour effects. Such models can be used to sulate the scope of the aberrations found in patients.
evaluate ICIs and other immunotherapies. This issue is further complicated by the fact that most
Finally, osteosarcomas arise spontaneously in tumour-​profiling panels include only subsets of genes
canines and have similar patterns of DNA alterations that do not encompass the entire pathway associated
to their human counterpart55,56. Veterinary clinical tri- with a given alteration. Tumours likely to be suscepti-
als in canines are feasible through comparative oncol- ble to a potential targeted therapy might therefore be
ogy consortiums sponsored by the NCI and other missed with the use of targeted gene panels. Hence, the
organizations57. Such trials bear some of the same issues emerging concept is that precision medicine for patients
as human clinical trials in that the canines require with osteosarcoma will require a broader view of cellular
informed consent from their owners for participation. pathway alterations (rather than of specific gene muta-
However, the owners might have differing thresholds of tions) to guide biomarker-​based clinical trials designed
acceptance for the toxicities associated with treatment to maximize the potential benefits of targeted therapies.
compared with parents of children with osteosarcoma. Although we have highlighted the weaknesses of each of
A potential major disadvantage of conducting these trials the model systems, we want to emphasize that these tools
is that the cost of the medications is often consigned to provide a crucially important starting point for develop-
the dog owners. ing hypotheses regarding which treatments will be active
Preclinical testing of potential targeted therapies and should be prioritized for clinical development.
should be considered in the context of pathway alter-
ations present in the PDX models. Owing to the inter- Targeted therapy of osteosarcoma
patient heterogeneity, if administered in an unbiased Tailoring therapy using a biomarker that predicts a
fashion across all models, a targeted therapy by defini- response (or resistance) to a specific molecularly tar-
tion will only induce responses in a subset of the mod- geted treatment is the central concept of precision med-
els tested. Thus, genomic characterization of the PDX icine. In the context of osteosarcoma, the challenges

www.nature.com/nrclinonc

0123456789();:
Reviews

facing precision medicine include the facts that most of lenvatinib (4-​month PFS of 33%)63 and pazopanib
the recurrent molecular alterations are difficult to tar- (median PFS of 5.5 months in patients with cancers of
get therapeutically (for example, those in TP53 or RB1) mixed histologies, 50% of which were osteosarcomas)64.
and that the alterations constituting dependencies of the These findings suggest a potential class effect despite the
tumour are often unclear on the background of multiple varied affinity of these compounds for different RTKs,
secondary genomic alterations. Moreover, clinical tri- with VEGFR being notably a common target of all of
als using pathway alterations as biomarkers for patient these TKIs, and have generated excitement in the field.
selection present a substantial challenge in a rare disease However, different end points were used to define ther-
with approximately 400 patients diagnosed annually in apeutic activity in some of the trials, particularly the
the USA. However, the COG analysis16 data, revealing older studies, which has complicated cross-​trial compar-
that the outcomes of patients with relapsed, unresectable isons to evaluate which agents to prioritize for further
osteosarcoma have remained unchanged across different evaluation.
phase II trials of numerous agents performed over a long Investigators have started to consider how to com-
period of time (with an aggregate 4-​month EFS of only bine TKIs with other agents to augment their activity.
12%), justifies the use of historical control groups. This The toxicities associated with combining TKIs with
situation has increased the feasibility of clinical trials chemotherapy are a major concern. Preliminary results
designed to evaluate the efficacy of novel therapies in with the combination of lenvatinib with ifosfamide
small subsets of patients with this rare disease. In multi- and etoposide demonstrated two dose-​limiting toxici-
ple single-​arm phase II trials performed by the COG, the ties (grade 4 thrombocytopenia and grade 3 bleeding
primary end point has been a 40% disease control rate in one patient, and grade 2 oral dysaesthesia, grade 2
(DCR) at 4 months43,44. Any active agents that demon- back pain and grade 1 muscle spasms in another) when
strate this level of activity will subsequently be tested lenvatinib was used at the recommended phase II
as a randomized addition to standard chemotherapy in single-​agent dose of 14 mg/m2 (ref.63). No efficacy data
patients with newly diagnosed osteosarcoma. This strat- were reported for this combination and this trial is ongo-
egy is predicated on the hypothesis that improvements in ing (NCT02432274; Supplementary Table 2). Building
frontline therapy have the highest likelihood of increas- on their initial promising experience with single-​agent
ing survival, with salvage chemotherapy thus far adding sorafenib59, Grignani et al.65 performed a phase II trial
no obvious benefit beyond surgery alone in patients with of sorafenib in combination with everolimus in patients
relapsed disease10–12. This strategy has enabled the rapid with relapsed, unresectable high-​grade osteosarcoma.
completion of clinical studies. For example, the phase II The primary end point of PFS ≥50% at 6 months was
AOST1322 trial of eribulin in patients with relapsed oste- not met, although the 6-​month PFS was 45%, with
osarcoma completed enrolment within a few months44,58. 66% of patients requiring dose reductions and/or short
Furthermore, studies performed worldwide have used treatment interruptions owing to toxicities65.
the 4-​month DCR end point, thus facilitating cross-​study Some important concerns exist regarding the combi-
comparisions16. The success of this approach underscores nation of multiple potentially active therapies. First, defin-
the potential for studies in biomarker-​selected subsets of ing an appropriate end point for the combination therapy
patients. Perhaps, however, some conceptual evolution of a novel agent with a known active chemotherapy is a
of the precision medicine paradigm is needed in osteosar- major challenge. Whether the end point typically applied
coma: rather than identifying direct target–drug relation- for single agents with unknown activity (4-month DCR
ships that define therapeutic activity, biomarker-​based of 40%) can be extrapolated to combinations of agents
selection could be used to increase the probability that with known activity is unclear. Second, the addition
an agent will be efficacious in a heterogeneous patient of the TKI might affect the administration of effective
population. In its simplest form, this approach might therapeutic agents. Owing to these challenges, ongo-
involve enrichment for patients with high-​risk disease, ing clinical trials are evaluating TKIs as maintenance
with MYC overexpression being a leading candidate therapy after the completion of planned chemother-
molecular biomarker of prognosis49,50. apy, including the placebo-​controlled REGOSTA trial
of regorafenib in patients with newly diagnosed bone
Tyrosine kinase inhibitors. Multiple receptor tyro­ sarcomas (NCT04055220; Supplementary Table 2).
sine kinases (RTKs) have been demonstrated to be Indeed, an emphasis in drug development for oste-
expressed in osteosarcoma cells. Most tyrosine kinase osarcoma has been placed on identifying active agents
inhibitors (TKIs) cannot necessarily be considered pre- with non-​overlapping toxicities that might facilitate the
cision medicines given that they act on multiple kinases combination with drugs used as standard treatments.
to a variable degree (Supplementary Table 2). Indeed, Restricting drug development to certain novel classes
clinical trials of TKIs in patients with osteosarcoma to of agents with non-​overlapping toxicities or at least to
date have not involved tumour profiling and thus the agents that are unlikely to necessitate changes to the
results do not provide a clear biological rationale for dose and intensity of the existing treatment and that
the activity observed. Nevertheless, multiple TKIs have also demonstrate single-​agent activity in patients with
demonstrated efficacy in patients with relapsed and/or relapsed osteosarcoma might be a more appropriate
unresectable disease, including sorafenib (4-​month strategy. The COG has been developing a randomized
progression-​free survival (PFS) of 46%)59, regorafenib trial testing the incorporation of cabozantinib into
(median PFS of 3.6 months versus 1.7 months with the upfront treatment of patients with osteosarcoma.
placebo)60, cabozantinib (4-​month PFS of 71%)61,62, Notably, the involvement of cooperative groups is

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

crucial to ensuring the timely completion of such studies PDX models39, cell lines and patient samples74. The inhi-
given the rarity of the disease and the large number of bition of CDK4/6 with palbociclib results in growth
patients needed. This requirement further supports the inhibition in cell lines expressing high levels of CDK4
stringency in agent selection. in vitro74. In vivo, single-​agent palbociclib induces
SRC is a non-​receptor tyrosine kinase involved in moderate tumour regression in a cisplatin-​resistant
multiple signalling pathways, including the mTOR and PDX model but greater tumour regressions in com-
MEK–ERK pathways, and is activated in the majority bination with sorafenib (as compared to either agent
of osteosarcomas66. Dasatinib, a kinase inhibitor with administered as monotherapy)75. AURKA/B have also
activity against SRC and ABL1, demonstrated tumour been demonstrated to be overexpressed in osteosar-
growth inhibition in two of six osteosarcoma PPTC coma patient samples76. The inhibition of AURKA led
PDX models, although no objective responses were to a maintained complete response (CR) in one of six
observed67. Moreover, a trial of saracatinib, a highly PDX models77 (Fig. 2); the inhibition of AURKB inhib-
selective inhibitor of SRC and ABL, was closed to accrual ited osteosarcoma cell growth in vitro and in vivo78.
after futility analysis did not demonstrate a PFS or OS Disappointingly, a phase II trial of the AURKA inhibitor
benefit post-​metastasectomy in patients with recurrent alisertib demonstrated no responses among 10 patients
osteosarcoma localized to the lung68. with osteosarcoma79. AURKB inhibitors have not been
Similarly to SRC, PDGFR has been shown to be evaluated in clinical trials in patients with osteosarcoma.
overexpressed in osteosarcomas, which has been asso- Dinaciclib, an inhibitor of CDK2, has been demon-
ciated with an unfavourable prognosis69. However, treat- strated to induce apoptosis in osteosarcoma cell lines
ment with imatinib, a TKI with activity against PDGFR, in vitro80; this agent induced stable disease in one of the
does not result in growth inhibition in preclinical mod- five PDX models tested81 (Fig. 2). Dinaciclib has not yet
els of osteosarcoma and was not efficacious in clinical been evaluated in clinical trials in the setting of osteo-
trials involving patients with this disease69,70. This raises sarcoma. These cell cycle targets require further study
the concern that detecting the target in the tumour to evaluate their potential role in the future treatment
might not suffice as a predictive biomarker and that of osteosarcoma.
other active pathways might negate target inhibition DDR pathways are important targets in osteosarcoma
through redundancy, compensatory changes and other given the larger numbers of somatic alterations observed
mechanisms of resistance. Such mechanisms are also in samples from patients22. As noted above, the somatic
likely to narrow the spectrum of tyrosine kinases that alterations are mostly chromosomal breaks and translo-
might confer true dependencies underlying the class cations rather than point mutations, indicative of defects
effect of multi-​target TKIs70. Nevertheless, selected TKIs in DDR mechanisms22,33. Currently, a clinical trial is eval-
are probably active agents in osteosarcoma, supporting uating the combination of the PARP inhibitor olaparib
the current approach of using small-​cohort phase II tri- with the ATR inhibitor ceralasertib in patients with
als with a high efficacy threshold to identify promising recurrent osteosarcoma (NCT04417062; Supplementary
novel therapies. Table 2) based on the rationale that the replication stress
induced by these agents will lead to mitotic lethality in
Targeting the cell cycle and DNA repair. Multiple subsets tumour cells that have already largely lost control of
of osteosarcomas are characterized by somatic altera- cell cycle checkpoints (owing to prevalent alterations in
tions affecting the cell cycle and/or DNA damage repair TP53 and RB1). Defects in DDR pathways similarly sug-
(DDR) pathways (Fig. 2) and clinical trials are being gest the possibility for clinical trials of monotherapy with
designed to evaluate precision medicine approaches these agents in biomarker-​selected patient populations.
predicated on such aberrations. For example, TP53 Alterations in the ATR pathway constitute one potential
aberrations compromise the G1 cell cycle checkpoint, biomarker for the sensitivity to treatments targeting the
thus increasing the dependence of tumour cells on the DDR in osteosarcoma82; however, this strategy needs to
G2 checkpoint to maintain DNA integrity and com- be validated in prospective clinical trials.
plete cell division. Therefore, agents that disrupt the G2
checkpoint, such as WEE1 inhibitors, might enhance Suppressing metastasis. Metastasis, most commonly
the activity of DNA-​damaging agents and induce the to the lungs, is a key determinant of the lethality of
mitotic lethality of TP53-​mutant osteosarcoma cells. osteosarcoma3. Substantial research has therefore been
Monotherapy with the WEE1 inhibitor AZD1775 has focused on the development of agents to disrupt the
limited activity in preclinical models of osteosarcoma; metastatic process. Such efforts have largely been driven
however, efficacy is observed in some of the models by the ‘seed and soil’ theory of metastasis, which posits
when this agent is combined with the topoisomerase 1 that a favourable microenvironment is required to create
inhibitor irinotecan71. Similarly, combining WEE1 inhib- a niche for the osteosarcoma metastases to take root. In
itors with gemcitabine and radiation results in enhanced particular, FAS signalling and macrophages have been
efficacy in PDX models of osteosarcoma72,73. These data identified as key factors mediating the formation of
highlight a potential mechanism to exploit the TP53 osteosarcoma metastases83,84. The role of macrophages
aberrations that are prevalent in osteosarcomas. is discussed further in the following section of this
CDK2, CDK4/6 and Aurora kinases (AURKA and/or Review. With regard to FAS, gemcitabine and entinostat
AURKB) have been identified as other potential molec- can augment the expression of this apoptotic receptor
ular targets involved in the cell cycle (Fig. 2). CDK4 has in osteosarcoma lung nodules and induce the regression
been demonstrated to be overexpressed in osteosarcoma of these lesions in preclinical models85,86. These findings

www.nature.com/nrclinonc

0123456789();:
Reviews

have laid the foundation for an ongoing clinical trial end point of OS, leading to the approval of this agent
evaluating aerosolized gemcitabine (NCT03093909; by the EMA but not the FDA5. Subsequent studies have
Supplementary Table 2). IL-6 and CXCL8 have also demonstrated that mifamurtide is well tolerated and is
been identified as signals mediating osteosarcoma associated with a 1-​year and 2-​year OS of 71.7% and
homing to the lungs and the combined inhibition of 45.9%, respectively, in patients with resectable recur-
IL-6 and CXCL8 signalling abrogates the development rent and/or metastatic disease101. These outcomes were
of lung metastases in several models of osteosarcoma87. observed in a non-​randomized trial but exceed those
Similarly, CXCL13 induces osteosarcoma cell migration expected for children with recurrent and/or metastatic
and invasion in vitro88. osteosarcoma with surgery, with or without chemother-
More recently, the epigenomic profiling of osteosar- apy, suggesting that this agent provides a therapeutic
coma metastases has revealed that changes in the activity benefit. Mifamurtide is currently available through com-
of enhancers associated with several genes are neces- passionate use programmes in the USA. The disparate
sary to permit metastatic dissemination89. Preclinical responses by regulatory agencies continues to challenge
data indicate that such changes drive the expression the more widespread use of mifamurtide for the treat-
of distinct subsets of genes within the first 24 hours of ment of osteosarcoma, although an ongoing trial might
metastatic colonization and later during metastatic out- clarify the potential benefit of this agent (NCT03643133,
growth, while other genes are constitutively activated89. Supplementary Table 2).
These findings suggest that phenotypic plasticity and Alternative approaches to augment immune
adaptation is required throughout various stages of the responses have been investigated. For example, in the
metastatic process. Notably, pharmacological inhibition phase III EURAMOS-1 trial, the addition of IFNα2b
of the epigenetic reader BET or functional inhibition of as a maintenance therapy was evaluated in patients
individual genes activated by the enhancers, such as with a good pathological response to preoperative
that encoding coagulation factor III/tissue factor (F3), MAP chemotherapy102; those with a poor pathologi-
suppressed lung metastasis in models of osteosarcoma89. cal response were randomly assigned to receive either
Evidence indicates that the lung microenvironment also standard postoperative MAP or MAP plus ifosfamide
needs to be altered in order to permit the invasion and and etoposide (MAP-​IE)6. Although EURAMOS-1 was
growth of osteosarcoma, which might provide avenues an achievement in that it was an international study and
for novel treatments90,91. the largest prospective trial ever performed in patients
with osteosarcoma, neither the addition of IFNα2b nor
Immunotherapy for osteosarcoma ifosfamide and etoposide improved EFS6,102.
An immune component of the treatment of osteosar- Novel strategies to specifically activate TAMs
coma has been envisaged since William Coley first are being investigated, including inhibition of the
observed tumour regressions following treatment with CD47–SIRPα signalling pathway. CD47 acts as an
bacterial toxins92. This perspective has been supported immune checkpoint by inhibiting the phagocytosis
by circumstantial evidence indicating that patients of non-​malignant cells by binding to SIRPα on mac-
who develop surgical site infections following primary rophages. Tumour cells can co-​opt this mechanism as
tumour resection have better outcomes93,94. Analyses of a means of immune evasion, and inhibitory anti-​CD47
the tumour microenvironment (TME) of osteosarco- monoclonal antibodies (mAbs) are under clinical devel-
mas consistently demonstrate an immune cell infiltrate opment as anticancer agents103. Few responses were
consisting of both macrophages and T cells95–98. The observed in an initial phase I trial of the first-​in-​class
presence of tumour-​infiltrating lymphocytes (TILs) anti-​CD47 mAb magrolimab in patients with various
has led to the hypothesis that ICIs could be effective in advanced-​stage cancers103; however, this agent was very
patients with this disease. In addition, osteosarcoma well tolerated, which might increase the feasibility of
was one of the first diseases in which the activation of using it as an adjunct therapy to improve the efficacy
tumour-​associated macrophages (TAMs) was pursued of other immunotherapies. With regard to osteosar-
as a treatment strategy. coma, CD47 is typically expressed at higher levels in
the tumour cells than in the surrounding tissue, and
Targeting macrophages. Early studies with muramyl antagonistic antibodies to CD47 have been shown to
tripeptide phosphatidylethanolamine (MTP-​PE; also increase macrophage-​mediated phagocytosis in vitro
known as mifamurtide) revealed that this agent can and to decrease the development of pulmonary metas-
activate blood monocytes to selectively kill tumour tases in PDX models104. Clinical trials are currently being
cells but not non-​malignant cells83,99. Mifamurtide can designed to evaluate whether inhibiting CD47 augments
activate the innate immune system via the pattern-​ the activity of other immunotherapies in patients with
recognition receptor NOD2, which is highly expressed in osteosarcoma. In one such proposed trial, an anti-​CD47
monocytes100. The initial preclinical and clinical experi- antibody will be combined with an anti-​GD2 antibody105.
ence with mifamurtide led to a phase III trial evaluating Lung macrophages have also been implicated as fac-
the addition of this agent to standard chemotherapy for tors that foster a stable niche for osteosarcoma metas-
patients with resectable non-​metastatic osteosarcoma, tases. Vascular cell adhesion molecule 1 (VCAM1) has
which was complicated by its factorial design involving been demonstrated to be expressed on tumour cells and
the parallel randomized testing of ifosfamide. Ultimately, is associated with an increased ability to metastasize106.
mifamurtide did not meet the primary EFS end point Indeed, VCAM1 expressed by osteosarcoma cells
but did demonstrate benefit according to the co-​primary can interact with the integrins expressed on lung

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

macrophages, which promotes cell migration and the col- (PR) and 5 (33.3%) had stable disease; the 6-​month PFS
onization of the pre-​metastatic niche88. Researchers are was 13.3%116. In the phase II SARC study of pembroliz­
currently developing antagonistic anti-​VCAM1 mAbs umab in patients with advanced-​stage sarcomas, only
to abrogate this interaction with the goal of reducing 1 (4.5%) of 22 patients with osteosarcoma enrolled had
pulmonary metastases107. a PR, with the majority discontinuing therapy owing to
disease progression117. The COG has also conducted a
Harnessing the immune infiltrate. In addition to TAMs, phase I/II trial of nivolumab in paediatric patients with
TILs also have an important role in the immune response relapsed solid tumours including 13 patients with oste-
to osteosarcoma, with ~75% of tumours expressing cell-​ osarcoma, all of whom had rapid disease progression
surface markers consistent with T cell and macrophage during treatment with this agent118. These results might
infiltrates98. However, an increased abundance of TILs is reflect fundamental differences between the biology
correlated with the expression of PD-​L1 and the related of paediatric and adult malignancies, in particular in
protein HHLA2 in osteosarcomas, which are associated terms of tumour mutational burden and potential neo-
with an unfavourable EFS95,96. By contrast, an increased antigen expression. Osteosarcoma has one of the high-
ratio of CD8+ cytotoxic T cells to FOXP3+ regulatory est tumour mutational burdens of any paediatric cancer
T cells in the TME has been associated with an improved type119; however, unlike most malignancies that occur
prognosis108. Further stratification revealed that nearly in adults, these mutations manifest as chromosomal
100% of patients with a CD8+ to FOXP3+ T cell ratio rearrangements rather than as point mutations37,119.
greater than the median were alive beyond 16 years Moreover, the majority (>70%) of genomic events
compared with <50% for those with a ratio less than the observed occur in non-​coding regions. Tumours with a
median108. Together, these data highlight the importance high predicted neoantigen expression also have high lev-
of tumour cell evasion of the immune infiltrate in the els of nonsense-​mediated decay factors, although such
pathogenesis of osteosarcoma. Accordingly, numerous tumours typically have higher immune scores37.
approaches are being taken to alleviate the immunosup- Despite these disappointing results, the biology of
pressive microenvironment of tumours, in particular immune checkpoints in osteosarcoma continues to drive
with ICIs or adoptive cell therapies. interest in this treatment approach. In mouse models
One approach to harnessing the immune system of this disease, PD-1 inhibition has been demonstrated
that is currently under investigation in osteosarcoma to decrease lung metastasis via the conversion of mac-
involves harvesting TILs from patient surgical speci- rophages from a pro-​tumour M2 phenotype into an
mens. These TILs are then expanded ex vivo, infused antitumour M1 phenotype120. These findings provide
back into the patient and activated in vivo through the a potential rationale for further clinical trials aim-
post-​infusion administration of IL-2 (NCT03449108)109. ing to improve the responses to ICIs (Supplementary
This approach is antigen agnostic, with the presumption Table 2). Avelumab, an mAb targeting PD-​L1 rather than
that these TILs, given their numbers and extrinsic acti- PD-1, is being evaluated in a multicentre phase II trial
vation, can overwhelm the immunosuppressive mecha- in patients with recurrent or progressive osteosarcoma
nisms of osteosarcomas and induce tumour regression. (NCT03006848). In an alternative approach, azacitidine
Similarly, the adoptive transfer of natural killer (NK) is being explored in patients with resectable relapsed
cells is another non-​specific mechanism of harness- osteosarcoma with the goal of increasing neoanti-
ing the immune system. NK cells are part of the innate gen expression and thereby increasing the efficacy of
system and can recognize malignant cells through a sys- nivolumab (NCT03628209)121. The COG is currently in
tem of activatory and inhibitory receptors; these cells the planning stages of a trial using radiation as another
have demonstrated preclinical activity against osteo- mechanism of increasing neoantigen expression as well
sarcoma both in vitro and in vivo110–112. Clinical trials as inducing other immunological changes and augment-
designed to evaluate the efficacy of adoptive NK cell ing responses to ICIs122. ICIs are also being combined
therapy in patients with osteosarcoma are under way with the anti-​CD73 antibody oleclumab in a study
(NCT03420963 and NCT02100891; Supplementary involving patients with advanced-​stage osteosarcoma
Table 2). Moreover, newer iterations of NK cell therapy (NCT04668300; Supplementary Table 2). This approach
analogous to the chimeric antigen receptor (CAR) T cell is predicated on the finding that tumours express-
approaches that are used in the treatment of B cell malig- ing CD73 accumulate extracellular adenosine, which
nancies create the opportunity to target specific antigens impairs antitumour T cell responses and thus constitutes
expressed by osteosarcoma cells113. another mechanism of immune evasion123; therefore, the
Members of the B7 family of immune-​checkpoint hope is that the combined inhibition of PD-1 or PD-​L1
proteins, including PD-​L1, HHLA2 and B7-​H3 (also and CD73 will increase the T cell response to osteosar-
known as CD276), have been demonstrated to be coma. B7-​H3 is also being explored as a therapeutic
expressed on the surface of osteosarcoma cells as a mech- target in osteosarcoma (as is discussed further below),
anism of immune evasion95,96,114,115. This observation has although using agents distinct from ICIs.
led to multiple clinical trials of ICIs targeting PD-1, the
receptor for PD-​L1, in patients with advanced-​stage Exploiting the osteosarcoma surfaceome
osteosarcoma, with somewhat disappointing results. A number of proteins are known to be enriched on the
For example, in the phase II PEMBROSARC trial of surface of osteosarcoma cells (Fig. 4), and research to dis-
pembrolizumab plus metronomic cyclophosphamide, cover additional clinically applicable cell-​surface targets
1 (6.7%) of 15 evaluable patients had a partial response is ongoing. Owing to the rarity of osteosarcoma, clinical

www.nature.com/nrclinonc

0123456789();:
Reviews

ADC (samrotamab
ADC vedotin)
(glembatumumab Positive preclinical CAR T cells
vedotin) results (EGFR806-EGFRt) ADCs (e.g.
Negative phase II Clinical activity in Phase I trial omburtamab,
trial phase I trial ongoing m276-PBD,
ADC MGC018 and
(trastuzumab Bi-specific DS-7300a)
mAb deruxtecan) T cell engager Positive preclinical
(trastuzumab) Phase II trial (Hu3F8-BsAb) results (m276-PBD)
Negative ongoing Phase I/II trial
phase II trial ongoing

GD2 LRRC15
GPNMB EGFR
HER2 B7-H3
mAb CAR T cells
Negative (e.g. GD2CART, CAR T cells
CAR T cells phase II trial iC9-GD2-CAR-VZV-CTL and
(HER2-CD28 (B7H3-EGFRt-DHFR)
(dinutuximab) GD2-CAR.OX40.28.z.ICD9) Positive preclinical
T cells) Phase I/II trial Phase I trials ongoing
Phase I trial results
ongoing Phase I trial ongoing
ongoing (Hu3F8)

Fig. 4 | Targeting the surfaceome of osteosarcoma. A number of cell-​surface molecules are commonly overexpressed on
osteosarcoma cells. Accordingly, a number of antibody-​based and/or cell therapy approaches targeting these molecules
are being studied for the treatment of osteosarcoma. For each approach, the stage of investigation or the results of trials
reported thus far are summarized. ADC, antibody–drug conjugate; CAR, chimeric antigen receptor; mAb, monoclonal
antibody.

trials investigating strategies to target components of the CR observed in two models127. Responses correlated
‘surfaceome’ of this disease have mostly used agents that with the tumoural level of LRRC15 expression128. Data
were developed for overlapping high-​priority targets in from the first-​in-​human phase I trial of samrotamab
other, more common malignancies. vedotin in 78 patients with various malignancies
demonstrated PRs in 4 (14.8%) and stable disease in
GPNMB. GPNMB has physiological roles in osteo- 8 (29.6%) of 27 patients with sarcoma, 10 of whom had
blast differentiation and osteoclast development and osteosarcoma129. The limited level of activity observed
has been demonstrated by immunohistochemistry to with samrotamab vedotin as well as with glembatu-
be expressed in 92.5% of osteosarcoma samples. The mumab vedotin might reflect the intrinsic activity of
GPNMB-​targeted ADC glembatumumab vedotin, which their MMAE payload: the PPTC results with microtu-
has the antimitotic microtubule inhibitor monomethyl bule inhibitors in PDX models of osteosarcoma con-
auristatin E (MMAE) as a payload, has demonstrated sistently demonstrate mixed responses, with some PDX
in vitro and in vivo preclinical activity124,125. However, in models being highly resistant to these agents (Fig. 3).
a phase II trial43, glembatumumab had limited activity Moreover, these responses, although better than those
against relapsed and/or refractory osteosarcoma (Fig. 4). observed with other agents (such as topoisomerase
Of the 22 patients enrolled, 1 patient had a PR (4.5%) inhibitors), have not been recapitulated clinically as best
and 2 patients had stable disease (9.1%)43. demonstrated by the lack of responses to eribulin in a
phase II trial involving patients with relapsed metastatic
LRRC15. Leucine-​ r ich repeat-​ c ontaining protein osteosarcoma44. Thus, reconstructing these ADCs with
15 (LRRC15) is a member of the leucine-​rich repeat a payload that has a greater efficacy in osteosarcoma,
superfamily of proteins, which have been implicated such as with pyrrolobenzodiazepine (PBD) or other
in cell adhesion, invasion and immune responses, DNA-​damaging agents, might increase their efficacy.
among other functions. Most osteosarcomas (91.3%) Nevertheless, consideration should also be given to the
have some degree of staining for LRRC15 on immuno­ importance of the target antigen.
histochemistry126. Samrotamab vedotin (also known
as ABBV-085) is an ADC comprising an anti-​LRRC15 HER2. HER2 is expressed in ~40% of osteosarcomas
mAb conjugated to MMAE (Fig. 4). Preclinically, samro- and has been heavily explored as a therapeutic target in
tamab vedotin resulted in the prolongation of EFS in five this disease, although with contradictory results130–132.
of seven PPTC PDX models of osteosarcoma in compar- In a trial with biomarker-​stratified treatment alloca-
ison with an isotype control ADC, with a maintained tion, the addition of anti-​HER2 mAb trastuzumab to

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

upfront MAP-​IE chemotherapy for patients with HER2-​ GD2-​targeted CAR T cell products as well as a Hu3FB-​
overexpressing metastatic osteosarcoma resulted in a based bispecific T cell-​engaging construct have been
similar 30-​month EFS and OS to that observed with developed and are currently being tested in early phase
MAP-​IE alone in those without HER2 overexpression clinical trials (Fig. 4; Supplementary Table 2).
(32% versus 32% and 59% versus 50%, respectively)133.
Whether HER2 is prognostic in osteosarcoma has been B7-​H3. B7-​H3 overexpression has been detected by
a controversial question; however, data derived from immunohistochemistry in 91.8% of osteosarcoma
233 patients enrolled on prospective clinical trials indi- specimens114. Unlike other immune-​checkpoint proteins,
cate that HER2 overexpression is not prognostic in terms antagonistic mAbs are not being used to directly inhibit
of EFS or OS131. In contrast with trastuzumab, HER2-​ B7-​H3 signalling and thereby activate an immune
targeted CAR T cell therapy has some activity against response but rather B7-​H3 is being exploited for the tar-
HER2+ recurrent and/or metastatic osteosarcoma, with geting of ADCs and CAR T cells towards osteosarcoma
3 of 14 evaluable patients having stable disease for between cells (Fig. 4). This strategy at least partially reflects the fact
12 and 15 weeks before undergoing tumour resection, 1 that the cellular function of B7-​H3 remains unclear142.
of whom had 90% tumour necrosis; these patients all The targeting of B7-​H3 was first explored in preclini-
remained in remission after 6, 12 and 16 months with no cal models of osteosarcoma and other cancers using the
further treatment134. Updated data from this trial indi- mAb omburtamab (8H9) conjugated to a Pseudomonas
cate that 1 patient has an ongoing CR for 32 months135. exotoxin143. Owing to systemic toxicities observed in
In addition, another patient with osteosarcoma had a PR these studies, intraperitoneal (NCT04022213) or intrathe­
lasting 9 months after receiving salvage chemotherapy cal (NCT03275402 and NCT00089245) administration
and a second dose of CAR T cells for initial progressive of this agent conjugated to the radioisotope iodine-131 is
disease134. Other agents with demonstrated activity even being evaluated in patients with other paediatric malig-
in tumours with low levels of HER2 expression, such nancies. B7-​H3-​targeted ADCs are also under devel-
as trastuzumab deruxtecan136, are also being explored opment, including m276-​PBD144,145. This agent carries
in patients with osteosarcoma (Fig. 4; Supplementary a DNA-​damaging agent PBD payload and induced
Table 2). Trastuzumab deruxtecan, an ADC with a topo­ CRs in two of five PDX models of osteosarcoma144,145.
isomerase inhibitor payload, prolonged the EFS in five of Another ADC targeting B7-​H3, MGC018, has a DNA-​
seven PDX models of osteosarcoma (as compared with alkylating payload (duocarmycin) and has demonstrated
vehicle controls)137. This work laid the foundation for a potent antitumour activity in mouse models of various
clinical trial of this agent in patients with osteosarcoma adult carcinomas146. This agent is currently undergoing
by the COG (NCT04616560). A potential limitation of evaluation in osteosarcoma PDX models by the PPTC.
HER2-​directed therapy relates to a lower prevalence and Preliminary data from the first-​in-​human phase I/II trial
more heterogeneous levels of HER2 expression in oste- of MGC018 (NCT03729596), which did not include
osarcomas as compared with the other target antigens patients with osteosarcoma, has revealed a manageable
discussed in this section. safety profile and evidence of activity in 4 of 20 patients
enrolled thus far147. DS-7300a, another B7-​H3-​targeted
GD2. GD2 is a disialoganglioside demonstrated to ADC, has a topoisomerase inhibitor payload and is cur-
be stably expressed on the surface of osteosarcoma rently being evaluated in a phase I/II trial in adults with
cells138,139. In humans, the expression of this glycosphin- advanced-​stage solid tumours (NCT04145622).
golipid in non-​malignant tissues is highly restricted, With regard to cell therapy strategies, B7-​H3-​targeted
mostly to the cerebellum and peripheral nerves, raising CAR T cells have demonstrated potent antitumour
the possibility for relatively specific tumour targeting. activity in one osteosarcoma xenograft model148. A
Dinutuximab (Ch14.18) is a chimeric mouse–human clinical trial of this CAR T cell product with a trun-
mAb targeting GD2 that can induce antibody-​dependent cated EGFR-​based safety switch is currently open to
cellular phagocytosis, antibody-​dependent cellular cyto- paediatric patients with osteosarcoma (NCT04483778;
toxicity and/or complement-​dependent cytotoxicity140. Supplementary Table 2). Selection of patients based on
Accordingly, GM-​CSF is typically used in conjunction the cell-​surface expression of B7-​H3 is not required
with this agent to help stimulate the anticancer activity of given the prevalence of this antigen in paediatric cancers.
TAMs. In a phase II trial of dinutuximab plus GM-​CSF
in patients with completely resected recurrent pulmo- EGFR. Anti-​EGFR CAR T cells are being investigated in
nary osteosarcoma, 12-​month EFS was 30.7%, which paediatric and young adult patients with osteosarcoma
did not meet the efficacy end point141. A phase II trial (NCT03618381; Supplementary Table 2). The major-
of humanized 3F8 (Hu3FB), another chimeric mAb ity of osteosarcoma cell lines (10 patient-​derived and
targeting GD2, plus GM-​CSF is currently ongoing in a 4 standard commercially available cell lines) express low
similar population of patients in second or later com- levels of EGFR149. Moreover, the EGFR TKI gefitinib
plete remission (NCT02502786; Supplementary Table 2). has activity against cell lines with high expression
The rationale for using these agents in the maintenance of EGFR only under conditions of serum starvation150.
setting is drawn from the experience with approved anti-​ EGFR has been found to be amplified in 28% of human
GD2 mAbs in neuroblastoma, another GD2-​expressing osteosarcomas151. A trial of EGFR-​targeted CAR T cells
paediatric tumour type, in which the greatest benefit is in patients with glioblastoma demonstrated trafficking
achieved when they are used for maintenance therapy of the cells to the tumour bed following intravenous
following initial disease control. In addition, several administration, with limited systemic toxicity; however,

www.nature.com/nrclinonc

0123456789();:
Reviews

analyses of post-​infusion resected specimens revealed disease. Several targets are currently being evaluated
the downregulation of cell-​surface EGFR expression and (Supplementary Table 2) with more coming down the
a more immunosuppressive TME152. pipeline. As discussed, these nascent strategies in oste-
osarcoma face several hurdles. First, the antigen needs
CAR T cell versus ADCs. CAR T cells and ADCs have to be of sufficient functional importance to avoid the
a similar specificity for the same target antigen; how- treatment-​related clonal selection of tumour cells with
ever, rather than requiring the antigen–antibody com- low or no expression of the target, particularly consider-
plex to deliver the payload, the CAR T cell approach ing the known intratumoural heterogeneity of osteosar-
exploits the intrinsic cytotoxicity machinery of T cells coma. Second, the optimal method of targeting, whether
to eliminate tumour cells. CAR T cells have the addi- with an mAb (typically requiring sufficient immune
tional benefit that they can persist in vivo for prolonged activation), an ADC (requiring an active payload) or a
periods of time, leading to continued eradication of cells cell therapy product (avoiding exhaustion), remains to
expressing a particular antigen153. CAR T cells might be determined for most if not all antigens and requires
also circumvent the challenge with ADCs relating to further clinical studies.
the requirement for a payload that is active against oste- Ultimately, novel agents will probably need to be
osarcoma. Nevertheless, CAR T cell therapy is associ- combined in order to overcome the intrinsic treatment
ated with concerns, including limited tumour homing, resistance observed in the osteosarcoma clinical trials
cytokine-​related toxicities, on-​target off-​tumour effects of the past. As noted in the example of lenvatinib63, the
and cellular exhaustion. NK cells incorporating the CAR high doses of cytotoxic chemotherapy currently used in
technology (CAR NK cells) are being explored as an patients with osteosarcoma leave limited space for addi-
alternative to overcome these potential drawbacks. For tional agents without prohibitive toxicities. Reducing
example, CAR NK cells do not seem to carry the same doses or the number of cycles might compromise the
risk of inducing cytokine storms and are able to survive outcomes achieved thus far. TKIs still remain the next
after killing multiple target cells, which in T cells can likely choice of agents to be incorporated with upfront
lead to exhaustion or activation-​induced cell death113. chemotherapy, whether as maintenance therapy, as
advocated by some who are concerned by their toxicities,
Conclusions or by developing novel strategies to safely integrate them
Over the past decade, the fruits of years of research using into the current chemotherapy regimens.
tissue banks, the development of preclinical models and The interest in immune-​based therapies is height-
genomic characterization have led to unprecedented ened by their different toxicity profiles, which do not
advances in our understanding of osteosarcoma. The necessarily overlap with known toxicities of cytotoxic
molecular characterization of osteosarcoma PDXs and chemotherapy. This scenario might be favourable for
patient samples have raised the possibility of designing integrating immunotherapies into current treatment
biomarker-​stratified clinical trials. Innovative approaches regimens if they are first found to be efficacious in
to drug development using small phase II trials with a clinical trials, similar to the strategy currently being
high bar for activity have provided the foundation for applied in clinical trials focused on paediatric haemato-
the rapid clinical evaluation of a multitude of novel logical malignancies (for example, NCT02166463 and
therapies. Furthermore, we recognize that new molec- NCT03914625).
ular techniques, such as single-​cell RNA sequencing to Immune-​based treatments might also have a role in
characterize tumour heterogeneity as well as the immune maintenance therapy, similar to that of anti-​GD2 mAbs
infiltrate and CRISPR or Sleeping Beauty screens for in patients with neuroblastoma. Indeed, these immuno­
novel therapeutic targets, will continue to enrich our therapies are likely to be most effective in the setting
understanding of osteosarcoma as they are applied to of minimal residual disease. Most patients with osteo-
this disease in the very near future, which will facilitate sarcoma who have disease recurrence have lung-​only
the prioritization of treatments that are most likely to metastasis, which probably occurs owing to the pres-
be effective154,155. The use of techniques such as multi- ence of micrometastases at the time of diagnosis. The
plexed ion beam imaging enables staining for 40 markers treatments outlined herein provide potential avenues
simultaneously in paraffin-​embedded tissue specimens, to eradicate these occult micrometastases before they
thus permitting rapid screening of patients for possible develop into overt metastatic disease. With all of the
enrolment in biomarker-​based trials156. Such a strategy is new biological discoveries, technologies, agents and
essential in conducting clinical trials of targeted therapies approaches, osteosarcoma seems to be a disease on the
in a very heterogeneous patient population. verge of marked advancement in patient outcomes after
The role of cell-​surface antigen expression in oste- more than four decades of stagnation.
osarcoma is an area that is clearly under active inves-
tigation with hopes of improving the treatment of the Published online xx xx xxxx

1. Mirabello, L., Troisi, R. J. & Savage, S. A. of 1,702 patients treated on neoadjuvant cooperative survival–a report from the Children’s Oncology Group.
Osteosarcoma incidence and survival rates from 1973 osteosarcoma study group protocols. J. Clin. Oncol. J. Clin. Oncol. 26, 633–638 (2008).
to 2004: data from the Surveillance, Epidemiology, 20, 776–790 (2002). 6. Marina, N. M. et al. Comparison of MAPIE
and End Results Program. Cancer 115, 1531–1543 4. Isakoff, M. S., Bielack, S. S., Meltzer, P. & Gorlick, R. versus MAP in patients with a poor response
(2009). Osteosarcoma: current treatment and a collaborative to preoperative chemotherapy for newly
2. Ottaviani, G. & Jaffe, N. The epidemiology of pathway to success. J. Clin. Oncol. 33, 3029–3035 diagnosed high-​grade osteosarcoma (EURAMOS-1):
osteosarcoma. Cancer Treat. Res. 152, 3–13 (2009). (2015). an open-​label, international, randomised
3. Bielack, S. S. et al. Prognostic factors in high-​grade 5. Meyers, P. A. et al. Osteosarcoma: the addition of controlled trial. Lancet Oncol. 17, 1396–1408
osteosarcoma of the extremities or trunk: an analysis muramyl tripeptide to chemotherapy improves overall (2016).

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

7. Ferrari, S. et al. Neoadjuvant chemotherapy with 30. Kovac, M. et al. Exome sequencing of osteosarcoma imbalance in canine osteosarcoma parallel the
methotrexate, cisplatin, and doxorubicin with or reveals mutation signatures reminiscent of BRCA human counterpart. Genes Chromosomes Cancer 50,
without ifosfamide in nonmetastatic osteosarcoma deficiency. Nat. Commun. 6, 8940 (2015). 859–874 (2011).
of the extremity: an Italian sarcoma group trial 31. Behjati, S. et al. Recurrent mutation of IGF signalling 56. Varshney, J., Scott, M. C., Largaespada, D. A. &
ISG/OS-1. J. Clin. Oncol. 30, 2112–2118 (2012). genes and distinct patterns of genomic rearrangement Subramanian, S. Understanding the osteosarcoma
8. Daw, N. C. et al. Frontline treatment of localized in osteosarcoma. Nat. Commun. 8, 15936 (2017). pathobiology: a comparative oncology approach.
osteosarcoma without methotrexate: results of the 32. Perry, J. A. et al. Complementary genomic approaches Vet. Sci. 3, 3 (2016).
St. Jude Children’s Research Hospital OS99 trial. highlight the PI3K/mTOR pathway as a common 57. Gordon, I., Paoloni, M., Mazcko, C. & Khanna, C.
Cancer 117, 2770–2778 (2011). vulnerability in osteosarcoma. Proc. Natl Acad. The comparative oncology trials consortium: using
9. Gaspar, N. et al. Results of methotrexate-​etoposide- Sci. USA 111, E5564–E5573 (2014). spontaneously occurring cancers in dogs to inform the
ifosfamide based regimen (M-​EI) in osteosarcoma 33. Wu, C. C. & Livingston, J. A. Genomics and the cancer drug development pathway. PLoS Med. 6,
patients included in the French OS2006/sarcome-09 immune landscape of osteosarcoma. Adv. Exp. e1000161 (2009).
study. Eur. J. Cancer 88, 57–66 (2018). Med. Biol. 1258, 21–36 (2020). 58. Isakoff, M. S. et al. Rapid protocol enrollment in
10. Daw, N. C. et al. Recurrent osteosarcoma with a single 34. Stephens, P. J. et al. Massive genomic rearrangement osteosarcoma: a report from the Children’s Oncology
pulmonary metastasis: a multi-​institutional review. acquired in a single catastrophic event during cancer Group. Pediatr. Blood Cancer 63, 370–371 (2016).
Br. J. Cancer 112, 278–282 (2015). development. Cell 144, 27–40 (2011). 59. Grignani, G. et al. A phase II trial of sorafenib in
11. Buddingh, E. P. et al. Prognostic factors in pulmonary 35. Cortés-​Ciriano, I. et al. Comprehensive analysis relapsed and unresectable high-​grade osteosarcoma
metastasized high-​grade osteosarcoma. Pediatr. Blood of chromothripsis in 2,658 human cancers using after failure of standard multimodal therapy: an Italian
Cancer 54, 216–221 (2010). whole-​genome sequencing. Nat. Genet. 52, 331–341 Sarcoma Group study. Ann. Oncol. 23, 508–516
12. Briccoli, A. et al. High grade osteosarcoma of the (2020). (2012).
extremities metastatic to the lung: long-​term results 36. Lorenz, S. et al. Unscrambling the genomic chaos 60. Davis, L. E. et al. Randomized double-​blind phase II
in 323 patients treated combining surgery and of osteosarcoma reveals extensive transcript fusion, study of regorafenib in patients with metastatic
chemotherapy, 1985–2005. Surg. Oncol. 19, recurrent rearrangements and frequent novel TP53 osteosarcoma. J. Clin. Oncol. 37, 1424–1431 (2019).
193–199 (2010). aberrations. Oncotarget 7, 5273–5288 (2016). 61. Smith, M. et al. Abstract LB-353: pediatric preclinical
13. Goorin, A. M. et al. Phase II/III trial of etoposide and 37. Wu, C. C. et al. Immuno-​genomic landscape of testing program (PPTP) stage 1 evaluation of
high-​dose ifosfamide in newly diagnosed metastatic osteosarcoma. Nat. Commun. 11, 1008 (2020). cabozantinib. Cancer Res. 73 (Suppl. 8), LB-353
osteosarcoma: a pediatric oncology group trial. 38. Lau, C. et al. The Genomic Landscape of Osteosarcoma: (2013).
J. Clin. Oncol. 20, 426–433 (2002). a Target Report. 2019 CTOS Annual Meeting 62. Italiano, A. et al. Cabozantinib in patients with
14. Berrak, S. G., Pearson, M., Berberoglu, S., Ilhan, I. E. (2019). advanced Ewing sarcoma or osteosarcoma (CABONE):
& Jaffe, N. High-​dose ifosfamide in relapsed pediatric 39. Sayles, L. C. et al. Genome-​informed targeted therapy a multicentre, single-​arm, phase 2 trial. Lancet Oncol.
osteosarcoma: therapeutic effects and renal toxicity. for osteosarcoma. Cancer Discov. 9, 46–63 (2019). 21, 446–455 (2020).
Pediatr. Blood Cancer 44, 215–219 (2005). 40. Northcott, P. A. et al. Medulloblastoma comprises 63. Gaspar, N. et al. Single-​agent expansion cohort of
15. Palmerini, E. et al. Gemcitabine and docetaxel in four distinct molecular variants. J. Clin. Oncol. 29, lenvatinib (LEN) and combination dose-​finding cohort
relapsed and unresectable high-​grade osteosarcoma 1408–1414 (2011). of LEN+etoposide (ETP)+ifosfamide (IFM) in patients
and spindle cell sarcoma of bone. BMC Cancer 16, 41. Houghton, P. J. et al. The pediatric preclinical testing (pts) aged 2 to ≤25 years with relapsed/refractory
280 (2016). program: description of models and early testing osteosarcoma (OS). J. Clin. Oncol. 36 (Suppl. 15),
16. Lagmay, J. P. et al. Outcome of patients with recurrent results. Pediatr. Blood Cancer 49, 928–940 (2007). 11527 (2018).
osteosarcoma enrolled in seven phase II trials through 42. Rokita, J. L. et al. Genomic profiling of childhood tumor 64. Aggerholm-​Pedersen, N., Rossen, P., Rose, H. &
Children’s Cancer Group, Pediatric Oncology Group, patient-​derived xenograft models to enable rational Safwat, A. Pazopanib in the treatment of bone
and Children’s Oncology Group: learning from the past clinical trial design. Cell Rep. 29, 1675–1689.e9 sarcomas: clinical experience. Transl Oncol. 13,
to move forward. J. Clin. Oncol. 34, 3031–3038 (2019). 295–299 (2020).
(2016). 43. Kopp, L. M. et al. Phase II trial of the glycoprotein 65. Grignani, G. et al. Sorafenib and everolimus for
17. Arndt, C. A. et al. Inhaled granulocyte-​macrophage non-​metastatic B-​targeted antibody-​drug conjugate, patients with unresectable high-​grade osteosarcoma
colony stimulating factor for first pulmonary glembatumumab vedotin (CDX-011), in recurrent progressing after standard treatment: a non-​
recurrence of osteosarcoma: effects on disease-​free osteosarcoma AOST1521: a report from the randomised phase 2 clinical trial. Lancet Oncol. 16,
survival and immunomodulation. a report from the Children’s Oncology Group. Eur. J. Cancer 121, 98–107 (2015).
Children’s Oncology Group. Clin. Cancer Res. 16, 177–183 (2019). 66. Urciuoli, E. et al. Src nuclear localization and its
4024–4030 (2010). 44. Isakoff, M. S. et al. A phase II study of eribulin in prognostic relevance in human osteosarcoma.
18. Biegel, J. A., Womer, R. B. & Emanuel, B. S. Complex recurrent or refractory osteosarcoma: a report from J. Cell Physiol. 233, 1658–1670 (2018).
karyotypes in a series of pediatric osteosarcomas. the Children’s Oncology Group. Pediatr. Blood Cancer 67. Kolb, E. A. et al. Initial testing of dasatinib by the
Cancer Genet. Cytogenet. 38, 89–100 (1989). 66, e27524 (2019). pediatric preclinical testing program. Pediatr. Blood
19. Bridge, J. A. et al. Cytogenetic findings in 73 45. Gill, J. et al. Dose-​response effect of eribulin in Cancer 50, 1198–1206 (2008).
osteosarcoma specimens and a review of the preclinical models of osteosarcoma by the pediatric 68. Baird, K. et al. Results of a randomized, double-​
literature. Cancer Genet. Cytogenet. 95, 74–87 preclinical testing consortium. Pediatr. Blood Cancer blinded, placebo-​controlled, phase 2.5 study of
(1997). 67, e28606 (2020). saracatinib (AZD0530), in patients with recurrent
20. Unni, K. K. & Dahlin, D. C. Osteosarcoma: pathology 46. Walkley, C. R. et al. Conditional mouse osteosarcoma, osteosarcoma localized to lung. Sarcoma 2020,
and classification. Semin. Roentgenol. 24, 143–152 dependent on p53 loss and potentiated by loss 7935475 (2020).
(1989). of Rb, mimics the human disease. Genes Dev. 22, 69. Kubo, T. et al. Platelet-​derived growth factor receptor
21. Dahlin, D. C. & Unni, K. K. Osteosarcoma of bone and 1662–1676 (2008). as a prognostic marker and a therapeutic target for
its important recognizable varieties. Am. J. Surg. 47. Berman, S. D. et al. Metastatic osteosarcoma induced imatinib mesylate therapy in osteosarcoma. Cancer
Pathol. 1, 61–72 (1977). by inactivation of Rb and p53 in the osteoblast lineage. 112, 2119–2129 (2008).
22. Chen, X. et al. Recurrent somatic structural variations Proc. Natl Acad. Sci. USA 105, 11851–11856 (2008). 70. Chugh, R. et al. Phase II multicenter trial of imatinib
contribute to tumorigenesis in pediatric osteosarcoma. 48. Jain, M. et al. Sustained loss of a neoplastic in 10 histologic subtypes of sarcoma using a bayesian
Cell Rep. 7, 104–112 (2014). phenotype by brief inactivation of MYC. Science 297, hierarchical statistical model. J. Clin. Oncol. 27,
23. Wang, L. L. et al. Augmented expression of MYC 102–104 (2002). 3148–3153 (2009).
and/or MYCN protein defines highly aggressive 49. Feng, W. et al. Myc is a prognostic biomarker and 71. Kolb, E. A. et al. Preclinical evaluation of the
MYC-​driven neuroblastoma: a Children’s Oncology potential therapeutic target in osteosarcoma. combination of AZD1775 and irinotecan against
Group study. Br. J. Cancer 113, 57–63 (2015). Ther. Adv. Med. Oncol. 12, 1758835920922055 selected pediatric solid tumors: a pediatric preclinical
24. Slayton, W. B., Schultz, K. R., Silverman, L. B. (2020). testing consortium report. Pediatr. Blood Cancer 67,
& Hunger, S. P. How we approach Philadelphia 50. Niu, J. et al. Identification of potential therapeutic e28098 (2020).
chromosome-​positive acute lymphoblastic leukemia targets and immune cell infiltration characteristics 72. Kreahling, J. M. et al. Wee1 inhibition by MK-1775
in children and young adults. Pediatr. Blood Cancer in osteosarcoma using bioinformatics strategy. leads to tumor inhibition and enhances efficacy of
67, e28543 (2020). Front. Oncol. 10, 1628 (2020). gemcitabine in human sarcomas. PLoS ONE 8,
25. Glover, J. et al. A summary of the osteosarcoma 51. Watanabe, A. et al. Osteosarcoma in Sprague-​Dawley e57523 (2013).
banking efforts: a report from the Children’s Oncology rats after long-​term treatment with teriparatide 73. PosthumaDeBoer, J. et al. WEE1 inhibition sensitizes
Group and the QuadW Foundation. Pediatr. Blood (human parathyroid hormone (1-34)). J. Toxicol. Sci. osteosarcoma to radiotherapy. BMC Cancer 11, 156
Cancer 62, 450–455 (2015). 37, 617–629 (2012). (2011).
26. Glover, J. et al. Osteosarcoma enters a post genomic 52. He, Y. et al. cFOS-​SOX9 axis reprograms bone 74. Zhou, Y. et al. Expression and therapeutic implications
era with in silico opportunities: generation of the High marrow-​derived mesenchymal stem cells into of cyclin-​dependent kinase 4 (CDK4) in osteosarcoma.
Dimensional Database for facilitating sarcoma biology chondroblastic osteosarcoma. Stem Cell Rep. 8, Biochim. Biophys. Acta Mol. Basis Dis. 1864,
research: a report from the Children’s Oncology Group 1630–1644 (2017). 1573–1582 (2018).
and the QuadW Foundation. PLoS ONE 12, e0181204 53. Zheng, B. et al. PD-1 axis expression in 75. Higuchi, T. et al. Sorafenib and palbociclib
(2017). musculoskeletal tumors and antitumor effect of combination regresses a cisplatinum-​resistant
27. Strauss, S. J. et al. Report from the 4th European nivolumab in osteosarcoma model of humanized osteosarcoma in a PDOX mouse model. Anticancer
Bone Sarcoma Networking meeting: focus on mouse. J. Hematol. Oncol. 11, 16 (2018). Res. 39, 4079 (2019).
osteosarcoma. Clin. Sarcoma Res. 8, 17 (2018). 54. Wagner, F. et al. Humanization of bone and bone 76. Tavanti, E. et al. Preclinical validation of Aurora
28. Wu, Z. L. et al. Development of a novel immune-​ marrow in an orthotopic site reveals new potential kinases-​targeting drugs in osteosarcoma. Br. J. Cancer
related genes prognostic signature for osteosarcoma. therapeutic targets in osteosarcoma. Biomaterials 109, 2607–2618 (2013).
Sci. Rep. 10, 18402 (2020). 171, 230–246 (2018). 77. Maris, J. M. et al. Initial testing of the aurora kinase A
29. Bousquet, M. et al. Whole-​exome sequencing in 55. Angstadt, A. Y. et al. Characterization of canine inhibitor MLN8237 by the pediatric preclinical testing
osteosarcoma reveals important heterogeneity of osteosarcoma by array comparative genomic program (PPTP). Pediatr. Blood Cancer 55, 26–34
genetic alterations. Ann. Oncol. 27, 738–744 (2016). hybridization and RT-​qPCR: Signatures of genomic (2010).

www.nature.com/nrclinonc

0123456789();:
Reviews

78. Zhao, Z. et al. Aurora B kinase as a novel molecular 100. Gisch, N., Buske, B., Heine, H., Lindner, B. & 122. Lhuillier, C. et al. Radiotherapy-​exposed CD8+ and
target for inhibition the growth of osteosarcoma. Zähringer, U. Synthesis of biotinylated muramyl CD4+ neoantigens enhance tumor control. J. Clin.
Mol. Carcinog. 58, 1056–1067 (2019). tripeptides with NOD2-stimulating activity. Invest. 131, e138740 (2021).
79. Mossé, Y. P. et al. A phase II study of alisertib in Bioorg. Med. Chem. Lett. 21, 3362–3366 (2011). 123. Zhang, B. CD73: a novel target for cancer
children with recurrent/refractory solid tumors or 101. Anderson, P. M. et al. Mifamurtide in metastatic and immunotherapy. Cancer Res. 70, 6407–6411 (2010).
leukemia: Children’s Oncology Group Phase I and Pilot recurrent osteosarcoma: a patient access study with 124. Roth, M. et al. Targeting glycoprotein NMB with
Consortium (ADVL0921). Clin. Cancer Res. 25, 3229 pharmacokinetic, pharmacodynamic, and safety antibody-​drug conjugate, glembatumumab vedotin,
(2019). assessments. Pediatr. Blood Cancer 61, 238–244 for the treatment of osteosarcoma. Pediatr. Blood
80. Fu, W. et al. The cyclin-​dependent kinase inhibitor (2014). Cancer 63, 32–38 (2016).
SCH 727965 (dinacliclib) induces the apoptosis 102. Bielack, S. S. et al. Methotrexate, doxorubicin, and 125. Kolb, E. A. et al. Initial testing (stage 1) of
of osteosarcoma cells. Mol. Cancer Ther. 10, cisplatin (MAP) plus maintenance pegylated interferon glembatumumab vedotin (CDX-011) by the pediatric
1018–1027 (2011). Alfa-2b versus MAP alone in patients with resectable preclinical testing program. Pediatr. Blood Cancer 61,
81. Gorlick, R. et al. Initial testing (stage 1) of the cyclin high-​grade osteosarcoma and good histologic 1816–1821 (2014).
dependent kinase inhibitor SCH 727965 (dinaciclib) response to preoperative MAP: first results of the 126. Cui, J. C. et al. Expression and clinical implications
by the pediatric preclinical testing program. Pediatr. EURAMOS-1 good response randomized controlled of leucine-​rich repeat containing 15 (LRRC15) in
Blood Cancer 59, 1266–1274 (2012). trial. J. Clin. Oncol. 33, 2279–2287 (2015). osteosarcoma. J. Orthop. Res. 38, 2362–2372
82. Li, X. et al. Inhibition of ATR-​Chk1 signaling blocks 103. Sikic, B. I. et al. First-​in-human, first-​in-class phase I (2020).
DNA double-​strand-break repair and induces trial of the anti-​CD47 antibody Hu5F9-G4 in patients 127. Hingorani, P. et al. ABBV-085, antibody-​drug conjugate
cytoplasmic vacuolization in metastatic osteosarcoma. with advanced cancers. J. Clin. Oncol. 37, 946–953 targeting LRRC15, is effective in osteosarcoma: a
Ther. Adv. Med. Oncol. 12, 1758835920956900 (2019). report by the Pediatric Preclinical Testing Consortium.
(2020). 104. Xu, J. F. et al. CD47 blockade inhibits tumor Mol. Cancer Ther. 20, 535–540 (2021).
83. Kleinerman, E. S., Murray, J. L., Snyder, J. S., progression human osteosarcoma in xenograft 128. Gill J. H. P. et al. Evaluation of ABBV-085, an
Cunningham, J. E. & Fidler, I. J. Activation of models. Oncotarget 6, 23662–23670 (2015). antibody-​drug conjugate targeting LRRC15, in
tumoricidal properties in monocytes from cancer 105. Theruvath, J. et al. Abstract PR07: GD2 is a osteosarcoma by the Pediatric Preclinical Testing
patients following intravenous administration macrophage checkpoint molecule and combined GD2/ Consortium. Connective Tissue Oncology Society
of liposomes containing muramyl tripeptide CD47 blockade results in synergistic effects and tumor Meeting (Poster). 137 (2019).
phosphatidylethanolamine. Cancer Res. 49, clearance in xenograft models of neuroblastoma and 129. Demetri, G. D. et al. First-​in-human phase 1 study
4665–4670 (1989). osteosarcoma. Cancer Res. 80 (Suppl. 14), PR07 of ABBV-085, an antibody-​drug conjugate (ADC)
84. Gordon, N. et al. Fas expression in lung metastasis (2020). targeting LRRC15, in sarcomas and other advanced
from osteosarcoma patients. J. Pediatr. Hematol. 106. Harjunpaa, H., Llort Asens, M., Guenther, C. & solid tumors. J. Clin. Oncol. 37, 3004–3004 (2019).
Oncol. 27, 611–615 (2005). Fagerholm, S. C. Cell adhesion molecules and their 130. Gorlick, R. et al. Expression of HER2/erbB-2 correlates
85. Gordon, N. & Kleinerman, E. S. The role of Fas/FasL roles and regulation in the immune and tumor with survival in osteosarcoma. J. Clin. Oncol. 17,
in the metastatic potential of osteosarcoma and microenvironment. Front. Immunol. 10, 1078 (2019). 2781–2788 (1999).
targeting this pathway for the treatment of 107. Huang, A. Y.-C. Targeting VCAM1-a4b1 signaling to 131. Gorlick, S. et al. HER-2 expression is not prognostic
osteosarcoma lung metastases. Cancer Treat. Res. ameliorate pulmonary osteosarcoma metastasis. NIH in osteosarcoma; a Children’s Oncology Group
152, 497–508 (2009). https://grantome.com/grant/NIH/R21-CA218790-01 prospective biology study. Pediatr. Blood Cancer 61,
86. Koshkina, N. V., Rao-​Bindal, K. & Kleinerman, E. S. (2017). 1558–1564 (2014).
Effect of the histone deacetylase inhibitor SNDX-275 108. Fritzsching, B. et al. CD8+/FOXP3+-ratio in 132. Gill J., Geller D., & Gorlick, R. in Current Advances in
on Fas signaling in osteosarcoma cells and the osteosarcoma microenvironment separates survivors Osteosarcoma (ed. Kleinerman, M. D. E. S.) 161–177
feasibility of its topical application for the treatment from non-​survivors: a multicenter validated (Springer International Publishing, 2014).
of osteosarcoma lung metastases. Cancer 117, retrospective study. Oncoimmunology 4, e990800 133. Ebb, D. et al. Phase II trial of trastuzumab in
3457–3467 (2011). (2015). combination with cytotoxic chemotherapy for
87. Gross, A. C. et al. IL-6 and CXCL8 mediate 109. Geukes Foppen, M. H., Donia, M., Svane, I. M. treatment of metastatic osteosarcoma with human
osteosarcoma-​lung interactions critical to metastasis. & Haanen, J. B. Tumor-​infiltrating lymphocytes for epidermal growth factor receptor 2 overexpression:
JCI Insight 3, e99791 (2018). the treatment of metastatic cancer. Mol. Oncol. 9, a report from the Children’s Oncology Group. J. Clin.
88. Liu, J. F. et al. CXCL13/CXCR5 interaction facilitates 1918–1935 (2015). Oncol. 30, 2545–2551 (2012).
VCAM-1-dependent migration in human 110. Guma, S. R. et al. Natural killer cell therapy and 134. Ahmed, N. et al. Human epidermal growth factor
osteosarcoma. Int. J. Mol. Sci. 21, 6095 (2020). aerosol interleukin-2 for the treatment of receptor 2 (HER2)-specific chimeric antigen receptor-​
89. Morrow, J. J. et al. Positively selected enhancer osteosarcoma lung metastasis. Pediatr. Blood Cancer modified T cells for the immunotherapy of HER2-
elements endow osteosarcoma cells with metastatic 61, 618–626 (2014). positive sarcoma. J. Clin. Oncol. 33, 1688–1696
competence. Nat. Med. 24, 176–185 (2018). 111. Kiany, S., Huang, G. & Kleinerman, E. S. Effect of (2015).
90. Murgai, M. et al. KLF4-dependent perivascular cell entinostat on NK cell-​mediated cytotoxicity against 135. Navai S. et al. Administration of HER2-CAR T cells
plasticity mediates pre-​metastatic niche formation osteosarcoma cells and osteosarcoma lung metastasis. after lymphodepletion safely improves T cell
and metastasis. Nat. Med. 23, 1176–1190 (2017). Oncoimmunology 6, e1333214 (2017). expansion and induces clinical responses in patients
91. Charan, M. et al. Tumor secreted ANGPTL2 facilitates 112. Tullius B. P., Setty B. A., Lee D. A. in Current Advances with advanced sarcomas (AACR Annual Meeting,
recruitment of neutrophils to the lung to promote in Osteosarcoma: Clinical Perspectives: Past, Present 2019).
lung pre-​metastatic niche formation and targeting and Future (eds Kleinerman E. S. & Gorlick R.) 136. Modi, S. et al. Antitumor activity and safety of
ANGPTL2 signaling affects metastatic disease. 141–154 (Springer International Publishing, 2020). trastuzumab deruxtecan in patients with HER2-low-​
Oncotarget 11, 510–522 (2020). 113. Habib, S., Tariq, S. M. & Tariq, M. Chimeric antigen expressing advanced breast cancer: results from
92. Coley, W. B. The treatment of inoperable sarcoma by receptor-​natural killer cells: the future of cancer a phase Ib study. J. Clin. Oncol. 38, 1887–1896
bacterial toxins (the mixed toxins of the Streptococcus immunotherapy. Ochsner J. 19, 186–187 (2019). (2020).
erysipelas and the Bacillus prodigiosus). Proc. R. Soc. 114. Wang, L. et al. B7-H3 is overexpressed in patients 137. Hingorani, P. et al. Abstract LB-217: preclinical
Med. 3, 1–48 (1910). suffering osteosarcoma and associated with tumor evaluation of trastuzumab deruxtecan (T-​DXd;
93. Jeys, L. M., Grimer, R. J., Carter, S. R., Tillman, R. M. aggressiveness and metastasis. PLoS ONE 8, e70689 DS-8201a), a HER2 antibody-​drug conjugate, in
& Abudu, A. Post operative infection and increased (2013). pediatric solid tumors by the Pediatric Preclinical
survival in osteosarcoma patients: are they associated? 115. McEachron, T. A., Triche, T. J., Sorenson, L., Testing Consortium (PPTC). Cancer Res. 80
Ann. Surg. Oncol. 14, 2887–2895 (2007). Parham, D. M. & Carpten, J. D. Profiling targetable (Suppl. 16), LB-217 (2020).
94. Chen, Y. U., Xu, S. F., Xu, M. & Yu, X. C. Postoperative immune checkpoints in osteosarcoma. Oncoimmunology 138. Roth, M. et al. Ganglioside GD2 as a therapeutic
infection and survival in osteosarcoma patients: 7, e1475873 (2018). target for antibody-​mediated therapy in patients
reconsideration of immunotherapy for osteosarcoma. 116. Le Cesne, A. et al. Programmed cell death 1 (PD-1) with osteosarcoma. Cancer 120, 548–554 (2014).
Mol. Clin. Oncol. 3, 495–500 (2015). targeting in patients with advanced osteosarcomas: 139. Poon, V. I. et al. Ganglioside GD2 expression is
95. Koirala, P. et al. Immune infiltration and PD-​L1 results from the PEMBROSARC study. Eur. J. Cancer maintained upon recurrence in patients with
expression in the tumor microenvironment are 119, 151–157 (2019). osteosarcoma. Clin. Sarcoma Res. 5, 4 (2015).
prognostic in osteosarcoma. Sci. Rep. 6, 30093 117. Tawbi, H. A. et al. Pembrolizumab in advanced 140. Keyel, M. E. & Reynolds, C. P. Spotlight on
(2016). soft-​tissue sarcoma and bone sarcoma (SARC028): dinutuximab in the treatment of high-​risk
96. Koirala, P. et al. HHLA2, a member of the B7 family, a multicentre, two-​cohort, single-​arm, open-​label, neuroblastoma: development and place in therapy.
is expressed in human osteosarcoma and is associated phase 2 trial. Lancet Oncol. 18, 1493–1501 (2017). Biologics 13, 1–12 (2019).
with metastases and worse survival. Sci. Rep. 6, 118. Davis, K. L. et al. Nivolumab in children and young 141. Hingorani, P. et al. Phase II study of
31154 (2016). adults with relapsed or refractory solid tumours or antidisialoganglioside antibody, dinutuximab, in
97. Wolf-​Dennen, K., Gordon, N. & Kleinerman, E. S. lymphoma (ADVL1412): a multicentre, open-​label, combination with GM-​CSF in patients with recurrent
Exosomal communication by metastatic osteosarcoma single-​arm, phase 1-2 trial. Lancet Oncol. 21, osteosarcoma (AOST1421): a report from the
cells modulates alveolar macrophages to an M2 541–550 (2020). Children’s Oncology Group. J. Clin. Oncol. 38
tumor-​promoting phenotype and inhibits tumoricidal 119. Gröbner, S. N. et al. The landscape of genomic (Suppl. 15), 10508 (2020).
functions. Oncoimmunology 9, 1747677 (2020). alterations across childhood cancers. Nature 555, 142. Picarda, E., Ohaegbulam, K. C. & Zang, X. Molecular
98. Corre, I., Verrecchia, F., Crenn, V., Redini, F. & Trichet, 321–327 (2018). pathways: targeting B7-H3 (CD276) for human cancer
V. The osteosarcoma microenvironment: a complex 120. Dhupkar, P., Gordon, N., Stewart, J. & Kleinerman, E. S. immunotherapy. Clin. Cancer Res. 22, 3425–3431
but targetable ecosystem. Cells 9, 976 (2020). Anti-​PD-1 therapy redirects macrophages from an M2 (2016).
99. Kleinerman, E. S., Erickson, K. L., Schroit, A. J., to an M1 phenotype inducing regression of OS lung 143. Onda, M., Wang, Q. C., Guo, H. F., Cheung, N. K. &
Fogler, W. E. & Fidler, I. J. Activation of tumoricidal metastases. Cancer Med. 7, 2654–2664 (2018). Pastan, I. In vitro and in vivo cytotoxic activities of
properties in human blood monocytes by liposomes 121. Hong, Y. K. et al. Epigenetic modulation enhances recombinant immunotoxin 8H9(Fv)-PE38 against
containing lipophilic muramyl tripeptide. Cancer Res. immunotherapy for hepatocellular carcinoma. breast cancer, osteosarcoma, and neuroblastoma.
43, 2010–2014 (1983). Cell Immunol. 336, 66–74 (2019). Cancer Res. 64, 1419–1424 (2004).

Nature Reviews | Clinical Oncology

0123456789();:
Reviews

144. Kurmasheva, R. et al. Abstract C003: initial testing of 158. Fuchs, N. et al. Long-​term results of the co-​operative protocol (IOR/OS-3) with high-​dose methotrexate,
m276-PBD CD276 antibody-​drug conjugate in German-​Austrian-Swiss osteosarcoma study group’s intraarterial or intravenous cisplatin, doxorubicin,
preclinical models of pediatric cancers by the Pediatric protocol COSS-86 of intensive multidrug and salvage chemotherapy based on histologic tumor
Preclinical Testing Consortium (PPTC). Mol. Cancer chemotherapy and surgery for osteosarcoma response. Tumori 85, 458–464 (1999).
Ther. 18 (Suppl. 12), C003 (2019). of the limbs. Ann. Oncol. 9, 893–899 (1998). 171. Smeland, S. et al. Scandinavian Sarcoma Group
145. Seaman, S. et al. Eradication of tumors through 159. Bacci, G. et al. Neoadjuvant chemotherapy for Osteosarcoma Study SSG VIII: prognostic factors
simultaneous ablation of CD276/B7-H3-positive osteosarcoma of the extremity: long-​term results for outcome and the role of replacement salvage
tumor cells and tumor vasculature. Cancer Cell 31, of the Rizzoli’s 4th protocol. Eur. J. Cancer 37, chemotherapy for poor histological responders.
501–515.e8 (2017). 2030–2039 (2001). Eur. J. Cancer 39, 488–494 (2003).
146. Scribner, J. A. et al. Preclinical development of 160. Goorin, A. M. et al. Presurgical chemotherapy
MGC018, a duocarmycin-​based antibody-​drug compared with immediate surgery and adjuvant Acknowledgements
conjugate targeting B7-H3 for solid cancer. chemotherapy for nonmetastatic osteosarcoma: The work of R.G. is supported by The University of Texas MD
Mol. Cancer Ther. https://doi.org/10.1158/ Pediatric Oncology Group Study POG-8651. J. Clin. Anderson Cancer Center as the H. Grant Taylor, M.D., W.W.
1535-7163.Mct-20-0116 (2020). Oncol. 21, 1574–1580 (2003). Sutow, M.D. and Margaret P. Sullivan, M.D. Distinguished
147. Powderly, J. D. et al. Preliminary dose escalation 161. Chou, A. J. et al. Addition of muramyl tripeptide to Chair in Pediatrics. J.G. and R.G. both acknowledge support
results from a phase I/II, first-​in-human study of chemotherapy for patients with newly diagnosed from The Foster Foundation, Swim Across America,
MGC018 (anti-​B7-H3 antibody-​drug conjugate) in metastatic osteosarcoma: a report from the Children’s the Osteosarcoma Institute, the QuadW Foundation and the
patients with advanced solid tumors. J. Clin. Oncol. 38 Oncology Group. Cancer 115, 5339–5348 (2009). Barbara Epstein Foundation.
(Suppl. 15), 3071–3071 (2020). 162. Serra, M. et al. May P-​glycoprotein status be used to
148. Majzner, R. G. et al. CAR T cells targeting B7-H3, a stratify high-​grade osteosarcoma patients? Results Author contributions
pan-​cancer antigen, demonstrate potent preclinical from the Italian/Scandinavian Sarcoma Group 1 Both authors contributed to all aspects of the preparation of
activity against pediatric solid tumors and brain treatment protocol. Int. J. Oncol. 29, 1459–1468 this manuscript.
tumors. Clin. Cancer Res. 25, 2560–2574 (2019). (2006).
149. Hassan, S. E. et al. Cell surface receptor expression 163. Whelan, J. S. et al. Survival from high-​grade localised Competing interests
patterns in osteosarcoma. Cancer 118, 740–749 extremity osteosarcoma: combined results and The authors declare no competing interests.
(2012). prognostic factors from three European Osteosarcoma
150. Sevelda, F. et al. EGFR is not a major driver for Intergroup randomised controlled trials. Ann. Oncol. Peer review information
osteosarcoma cell growth in vitro but contributes to 23, 1607–1616 (2012). Nature Reviews Clinical Oncology thanks Bruno Fuchs,
starvation and chemotherapy resistance. J. Exp. Clin. 164. Link, M. P. et al. The effect of adjuvant chemotherapy Dominique Heymann, Massimo Serra and the other, anony-
Cancer Res. 34, 134 (2015). on relapse-​free survival in patients with osteosarcoma mous, reviewer(s) for their contribution to the peer review of
151. Huang, Z. et al. Clinicopathological and prognostic of the extremity. N. Engl. J. Med. 314, 1600–1606 this work.
values of ErbB receptor family amplification in (1986).
primary osteosarcoma. Scand. J. Clin. Lab. Invest. 79, 165. Marec-​Berard, P. et al. Methotrexate-​etoposide- Publisher’s note
601–612 (2019). ifosfamide compared with doxorubicin-​cisplatin- Springer Nature remains neutral with regard to jurisdictional
152. O’Rourke, D. M. et al. A single dose of peripherally ifosfamide chemotherapy in osteosarcoma treatment, claims in published maps and institutional affiliations.
infused EGFRvIII-​directed CAR T cells mediates patients aged 18–25 years. J. Adolesc. Young Adult
antigen loss and induces adaptive resistance in Oncol. 9, 172–182 (2020). Supplementary information
patients with recurrent glioblastoma. Sci. Transl Med. 166. Winkler, K. et al. Neoadjuvant chemotherapy for The online version contains supplementary material available
https://doi.org/10.1126/scitranslmed.aaa0984 osteogenic sarcoma: results of a Cooperative at https://doi.org/10.1038/s41571-021-00519-8.
(2017). German/Austrian study. J. Clin. Oncol. 2, 617–624
153. Schultz, L. Chimeric antigen receptor T cell therapy (1984).
for pediatric B-​ALL: narrowing the gap between early 167. Winkler, K. et al. Neoadjuvant chemotherapy of
Related links
Bone Cancer Research Trust: https://www.bcrt.org.uk/
and long-​term outcomes. Front. Immunol. 11, 1985 osteosarcoma: results of a randomized cooperative
Children’s Oncology Group (COG): https://
(2020). trial (COSS-82) with salvage chemotherapy based
childrensoncologygroup.org/index.php/
154. Zhou, Y. et al. Single-​cell RNA landscape of on histological tumor response. J. Clin. Oncol. 6,
childrens-​oncology-​group
intratumoral heterogeneity and immunosuppressive 329–337 (1988).
Count Me in: https://www.broadinstitute.org/count-me-in
microenvironment in advanced osteosarcoma. 168. Bramwell, V. H. et al. A randomized comparison of
euroBoNeT: https://cordis.europa.eu/project/id/18814
Nat. Commun. 11, 6322 (2020). two short intensive chemotherapy regimens in children
iTCC P4: https://www.itccp4.eu
155. Moriarity, B. S. et al. A Sleeping Beauty forward and young adults with osteosarcoma: results in
National Cancer institute (NCi) Therapeutically Applicable
genetic screen identifies new genes and pathways patients with metastases: a Study of the European
Research to Generate effective Treatments (TARGeT)
driving osteosarcoma development and metastasis. Osteosarcoma Intergroup. Sarcoma 1, 155–160
Osteosarcoma project: https://ocg.cancer.gov/programs/
Nat. Genet. 47, 615–624 (2015). (1997).
target/projects/osteosarcoma
156. Parra, E. R., Francisco-​Cruz, A. & Wistuba, I. I. 169. Le Deley, M. C. et al. SFOP OS94: a randomised trial
NCi Paediatric Preclinical Testing Consortium: http://www.
State-​of-the-​art of profiling immune contexture in comparing preoperative high-​dose methotrexate plus
ncipptc.org/
the era of multiplexed staining and digital analysis to doxorubicin to high-​dose methotrexate plus etoposide
Osteosarcoma Project: http://www.osproject.org
study paraffin tumor tissues. Cancers 11, 247 (2019). and ifosfamide in osteosarcoma patients. Eur. J.
Quadw Foundation: http://www.quadw.org/
157. Anninga, J. K. et al. Chemotherapeutic adjuvant Cancer 43, 752–761 (2007).
treatment for osteosarcoma: where do we stand? 170. Ferrari, S. et al. Nonmetastatic osteosarcoma of the
Eur. J. Cancer 47, 2431–2445 (2011). extremity: results of a neoadjuvant chemotherapy © Springer Nature Limited 2021

www.nature.com/nrclinonc

0123456789();:

You might also like