2023 SABCS Highlights

CE / CME

Highlights From the 2023 San Antonio Breast Cancer Symposium

Physician Assistants/Physician Associates: 1.25 AAPA Category 1 CME credits

Nurses: 1.25 Nursing contact hours

Physicians: maximum of 1.25 AMA PRA Category 1 Credits

Pharmacists: 1.25 contact hours (0.125 CEUs)

Released: February 23, 2024

Expiration: February 22, 2025

Sara A. Hurvitz
Sara A. Hurvitz, MD, FACP
Joyce O'Shaughnessy
Joyce O'Shaughnessy, MD

Activity

Progress
1 2
Course Completed

KEYNOTE-756 Subgroup Analysis: Neoadjuvant Pembrolizumab Plus Chemotherapy Followed by Adjuvant Pembrolizumab Plus Endocrine Therapy in High-Risk, ER-Positive/HER2-Negative EBC

Joyce O’Shaughnessy, MD:
We will start by discussing new analyses of the phase III KEYNOTE-756 and CheckMate 7FL trials. These studies are both evaluating perioperative ICI-based regimens in high-risk ER-positive/HER2-negative EBC.

The first analysis assessed pCR rates across patient subgroups in KEYNOTE-756.1 The first interim analysis of this trial was presented at ESMO 2023.2 In brief, KEYNOTE-756 enrolled 1278 patients with grade 3, stage II/III ER-positive/HER2 negative invasive ductal breast carcinoma. During the neoadjuvant phase, patients received pembrolizumab or placebo for 4 cycles plus paclitaxel for 12 weeks, followed by pembrolizumab or placebo plus either doxorubicin and cyclophosphamide or epirubicin and cyclophosphamide for 4 cycles. After surgery, patients received adjuvant pembrolizumab or placebo for 6 months, with endocrine therapy given for up to 10 years.

The coprimary endpoints are the rates for pCR, defined as ypT0/Tis ypN0, and event-free survival (EFS).

KEYNOTE-756 Subgroup Analysis: pCR at First Interim Analysis

Joyce O’Shaughnessy, MD:
At ESMO 2023, Cardoso and colleagues2 presented the results of the interim analysis, in which KEYNOTE-756 met its pCR endpoint. The pCR rate was 24.3% in the pembrolizumab arm vs 15.6% in the placebo arm—a statistically significant absolute difference of 8.5% (95% CI: 4.2%-12.8%; P = .00005).1

KEYNOTE-756 Subgroup Analysis: pCR at First Interim Analysis by Subgroup

Joyce O’Shaughnessy, MD:
On behalf of Cardoso and colleagues,1 I presented data on pCR rates across patient subgroups at SABCS 2023. The pCR benefit with the addition of pembrolizumab increased as PD-L1 expression increased. We see an absolute difference of 9.8% in those with a PD-L1 CPS ≥1, which increases to 13.2% with CPS ≥10 and 17.4% with CPS ≥20.

This “stairstep” relationship between benefit and PD-L1 CPS in ER-positive/HER2-negative EBC is particularly notable because these results contrast with those reported in early-stage TNBC. In the phase III KEYNOTE-522 trial, the absolute increase in the pCR rates did not vary by PD-L1 CPS in patients with early TNBC when pembrolizumab was added to neoadjuvant chemotherapy and then continued as monotherapy in the adjuvant phase.3 Patients with early TNBC and a PD-L1 CPS <1 had an absolute improvement in pCR rate of 18.3% with pembrolizumab, whereas those with a PD-L1 CPS ≥20 had an absolute improvement of 18.5%.

KEYNOTE-756 Subgroup Analysis: pCR at First Interim Analysis by Subgroup (Continued)

Joyce O’Shaughnessy, MD:
One of the more important clinical findings in this subgroup analysis of KEYNOTE-756 was the greater benefit in patients with a PD-L1 CPS ≥1 and ER expression <10%.1 In this subgroup, the addition of pembrolizumab was associated with an absolute difference in pCR rate of 24.2% (95% CI: 1%-45.1%) vs the addition of placebo. The pCR rate with pembrolizumab was 57.6%, which is closer to the pCR rate of 64.8% reported with pembrolizumab in early TNBC.3 By contrast, in KEYNOTE-756, the subgroup with a PD-L1 CPS <1 and ER expression ≥10% had an absolute difference in pCR rate of 4.6%.1

Another interesting finding was that patients derived more absolute benefit from receiving pembrolizumab when they had “full exposure” to the chemotherapy regimen, defined as 10-12 doses of paclitaxel weekly and all 4 cycles of anthracycline and cyclophosphamide, vs less than full exposure.1 Among those with full exposure, the pCR rate with placebo was 16.9% vs 26.2% with pembrolizumab, for an absolute difference of 9.3%. Among those with less than full exposure, the pCR rates were 6.4% vs 13.2%, respectively, for an absolute difference of 6.8%.

KEYNOTE-756 Subgroup Analysis: Immune-Mediated AEs in Neoadjuvant Phase

Joyce O’Shaughnessy, MD:
No new safety signals were observed in this trial.1

KEYNOTE-756 Subgroup Analysis: Clinical Implications

Joyce O’Shaughnessy, MD:
KEYNOTE-756 already demonstrated a significant pCR benefit with the addition of pembrolizumab to neoadjuvant chemotherapy and adjuvant endocrine therapy (without abemaciclib) in patients with grade 3 ER-positive/HER2-negative EBC in the intention-to-treat (ITT) population.2 This subgroup analysis observed that pCR benefit increased as PD-L1 CPS increased. Furthermore, there was a particularly pronounced pCR benefit in those with ER expression <10%.

Despite these promising results, we must wait for data from the other coprimary endpoint, EFS, before adopting this strategy in practice. However, I expect that some of these larger absolute benefits in pCR rate will translate into significantly improved EFS.

CheckMate 7FL Biomarker Analysis: Addition of Nivolumab vs Placebo to Neoadjuvant Chemotherapy and Adjuvant Endocrine Therapy in High-Risk ER-Positive/HER2-Negative EBC

Joyce O’Shaughnessy, MD:
Next, we will discuss an exploratory biomarker analysis of the phase III CheckMate 7FL trial, which I consider to be a “sister” trial to KEYNOTE-756. CheckMate 7FL enrolled 521 patients with high-risk ER-positive/HER2-negative EBC that was clinical stage II//III and either grade 3 with ER expression ≥1% or grade 2 with ER expression 1% to 10%.4 During the neoadjuvant phase, patients received nivolumab or placebo plus paclitaxel for 4 cycles, followed by nivolumab or placebo plus an anthracycline and cyclophosphamide for 4 cycles. After surgery, patients received adjuvant nivolumab or placebo plus endocrine therapy.

After abemaciclib received approval in the adjuvant setting, the trial design for CheckMate 7FL was amended in April 2022 out of concern that a CDK4/6 inhibitor could not be given safely with an anti–PD-1 agent. The primary endpoint was changed to only include pCR, defined as ypT0/Tis ypN0, and no further patients were enrolled after 521 participants were randomized. The 11 patients enrolled at the Russian site—which was closed because of the ongoing conflict in that region—were excluded from analysis, and the modified ITT population consisted of 510 patients.

CheckMate 7FL Biomarker Analysis: pCR and Residual Cancer Burden 0-1 Rates by PD-L1 Status per SP142 and 28-8 CPS Assays

Joyce O’Shaughnessy, MD:
Loi and colleagues5 first presented results from CheckMate 7FL at ESMO 2023, where they reported that the addition of nivolumab significantly improved the pCR rate by 10.5% (95% CI: 4.0%-16.9%; P = .0021) vs placebo in the modified ITT population. This absolute benefit in the pCR rate is similar to that reported with the addition of pembrolizumab in KEYNOTE-756.1

In the biomarker analysis presented at SABCS 2023, we saw again a “stairstep” relationship between increasing pCR benefit with an anti–PD-1 agent and increasing PD-L1 expression in the setting of ER-positive/HER2-negative EBC.4 Of interest, the absolute difference in pCR rate with nivolumab vs placebo was 52.3% in patients with PD-L1 CPS ≥20.

CheckMate 7FL Biomarker Analysis: pCR and Residual Cancer Burden 0-1 Rates by ER and Progesterone Expression in Grade 3 Tumors

Joyce O’Shaughnessy, MD:
This analysis also reported greater benefit in patients with grade 3 disease and ER expression ≤50%, where the absolute difference in pCR rate was 29.3% vs 7.4% in those with ER expression >50%.4

CheckMate 7FL: Clinical Implications

Joyce O’Shaughnessy, MD:
Taken together, these subgroup analyses of the KEYNOTE-759 and CheckMate 7FL trials indicate that we can identify patients with high-risk ER-positive/HER2-negative EBC who are more likely to benefit from the addition of an anti–PD-1 agent to neoadjuvant chemotherapy and adjuvant endocrine therapy. I believe we need to wait for EFS data from KEYNOTE-756 before integrating pembrolizumab into this setting in practice. Unfortunately, we will not be getting EFS data from the CheckMate 7FL trial because of the protocol amendment.

Sara A. Hurvitz, MD:
I also found these data to be very interesting. If an anti–PD-1 agent is approved for patients with hormone receptor–positive EBC, I believe these data will help select appropriate patients. I found it interesting that PD-L1 expression was both prognostic and predictive of response in ER-positive EBC, which we did not see in patients with early TNBC on the KEYNOTE-522 study, as you mentioned earlier.

Joyce O’Shaughnessy, MD:
I would speculate that this is related to the biologic continuum from the luminal A subtype, which is strongly driven by ER, to the luminal B subtype, which has greater genomic instability with more neoantigens.6 From my understanding, when a patient has aggressive disease where chemotherapy is increasing tumor antigenicity, the anti–PD-1 agent will be efficacious. However, patients can have grade 3 disease with strong ER and progesterone receptor positivity and still be relatively resistant to chemotherapy. To me, this is saying that we need chemotherapy to work to be able to see the benefit from the anti–PD-1 agent.

Based on recent analyses of the phase III KEYNOTE-756 and CheckMate 7FL trials, which of the following subgroups of patients with high-risk estrogen receptor (ER)‒positive/HER2-negative early breast cancer (EBC) is most likely derive the highest benefit from the addition of an immune checkpoint inhibitor (ICI) to neoadjuvant chemotherapy and adjuvant endocrine therapy?

NATALEE: Final iDFS Analysis of Adjuvant Ribociclib Plus an NSAI in Hormone Receptor–Positive/HER2-Negative EBC

Sara A. Hurvitz, MD:
Continuing our discussion of studies in the setting of hormone receptor–positive/HER2-negative EBC, Hortobagyi and colleagues7 presented the final invasive disease–free survival (iDFS) analysis of the open-label phase III NATALEE trial at SABCS 2023. NATALEE is comparing the addition of the CDK4/6 inhibitor ribociclib for 3 years to adjuvant endocrine therapy with a nonsteroidal aromatase inhibitor (NSAI) vs NSAI alone in 5101 patients with intermediate-/high-risk hormone receptor–positive/HER2-negative EBC. Of note, ribociclib was started at 400 mg daily in this trial—a lower dose than the 600 mg indicated for ABC.8

The final protocol-specified iDFS analysis was performed after approximately 500 events,7 updating the interim analysis of iDFS presented earlier in 2023.9

NATALEE: iDFS (Primary Endpoint)

Sara A. Hurvitz, MD:
With a median follow-up of 33.3 months, the 3-year iDFS rate was significantly improved with the addition of ribociclib to adjuvant NSAI vs NSAI alone.7 The 3-year iDFS rate was 90.7% with ribociclib plus NSAI vs 87.6% with NSAI alone, yielding an absolute improvement of 3.1% and a statistically significant HR of 0.749 (95% CI: 0.628-0.892; nominal 1-sided P = .0006). This absolute benefit in the 3-year iDFS rate is greater than the 1.5% benefit reported at 2 years.

NATALEE: iDFS in Key Patient Subgroups

Sara A. Hurvitz, MD:
I found the subgroup analyses of iDFS benefit to be particularly important. Recall that the monarchE trial evaluating adjuvant abemaciclib plus endocrine therapy enrolled patients with high-risk disease in this setting, whereas NATALEE permitted patients with node-negative stage II disease to enroll.7,10

The NATALEE investigators presented an informative breakdown of iDFS based on stage and nodal status.

Of interest, the HR—and therefore the iDFS benefit—with adjuvant ribociclib was fairly consistent for stage II vs III and node-positive vs node-negative disease. Of course, patients with more advanced disease had worse iDFS outcomes, but the absolute benefit from ribociclib was consistent across subtypes defined by stage and nodal status.

NATALEE: Other Survival Outcomes

Sara A. Hurvitz, MD:
With a median follow-up of 35.9 months for OS, these data were still quite immature, with <4% of events occurring in each arm.7

NATALEE: Safety

Sara A. Hurvitz, MD:
The safety outcomes in this final iDFS analysis were similar to those reported in the interim analysis.7,9 In terms of QT interval prolongation—an established part of the safety profile for ribociclib—it was reassuring to see that QT interval prolongation occurred at any grade in only 5.3% of patients and at grade ≥3 in 1.0% of patients who received adjuvant ribociclib. Liver-related adverse events (AEs) of any grade occurred in 26.4% of patients with ribociclib, with grade ≥3 liver-related AEs in 8.6%.

Discontinuations due to AEs occurred in 19.5% of patients who received ribociclib in the interim analysis. This rate increased by only 0.8% in the final iDFS analysis, with liver-related AEs being the most common reason.

NATALEE: Clinical Implications

Sara A. Hurvitz, MD:
I suspect that ribociclib may soon receive a new FDA-approved indication for the adjuvant setting. If approved, it will be interesting to see if the indication is for the entire patient population enrolled on NATALEE, which included patients with intermediate-risk and high-risk disease.

I am very interested in the long-term follow-up for NATALEE and monarchE to determine whether there is an OS benefit and whether the iDFS benefit continues to increase once all patients are off their protocol-specified therapy.

Joyce O’Shaughnessy, MD:
I agree that it is very promising to see that the absolute benefit in iDFS has been maintained and has even grown with further follow-up of NATALEE.

Nonetheless, we should bear in mind that approximately 20% of patients still have ongoing ribociclib treatment. It will be crucial to see outcomes for node-negative vs node-positive subgroups at 5 years, when all patients will have been off ribociclib for 2 years.

That being said, I find adjuvant ribociclib plus an NSAI to be promising. Assuming that this regimen is approved by the FDA and recommended by National Comprehensive Cancer Network guidelines, I anticipate using it for my patients who are not eligible for abemaciclib. This would include patients with higher-risk node‑negative disease or lower-risk node-positive disease.

monarchE: Molecular Profiling of Primary Tumor Tissue and ctDNA Detection in Patients With High-Risk, Node-Positive Hormone Receptor–Positive/HER2-Negative EBC Who Received Adjuvant ET With or Without Abemaciclib

Joyce O’Shaughnessy, MD:
Continuing our discussion of adjuvant CDK4/6 inhibition for EBC, we will next review 2 studies performed using data from the monarchE trial. In brief, monarchE is an ongoing phase III trial comparing the addition of abemaciclib for 2 years to adjuvant endocrine therapy vs endocrine therapy alone in 5637 patients with high-risk, node-positive hormone receptor–positive/HER2-negative EBC.10 This trial led to the FDA approval and adoption of adjuvant abemaciclib as the current standard of care in this setting.11,12 More recently, we saw 5-year outcomes presented at ESMO 2023 and subsequently published in 2024.13,14 These data showed that adding abemaciclib to adjuvant endocrine therapy has continued to benefit patients in this setting, with iDFS curves continuing to separate and an absolute benefit of 7.6% at 5 years.

At SABCS 2023, Turner and colleagues15 presented a molecular profiling study of primary tumor tissue, and Loi and colleagues16 presented results from a pilot study on circulating tumor DNA (ctDNA) detection in monarchE participants. We will first discuss the results of the molecular profiling study.

monarchE Molecular Profiling: Study Design

Joyce O’Shaughnessy, MD:
This analysis evaluated the treatment effect of abemaciclib in a biomarker population of 1400 monarchE participants consisting of, first, a stratified, random sample of the ITT population and, second, all patients with iDFS events at the primary analysis cutoff.15 Baseline primary tumors from this population were then analyzed using whole-exome sequencing (WES) and exome-capture RNA sequencing (RNAseq). The investigators then examined the association between iDFS and various biomarkers in participants who had received abemaciclib plus endocrine therapy or endocrine therapy alone.

We should note that the iDFS benefit with the addition of abemaciclib was consistent across the ITT population and the subpopulations analyzed using WES and RNAseq.

monarchE Molecular Profiling: iDFS Across Intrinsic Molecular Subtypes

Joyce O’Shaughnessy, MD:
The WES data were used to assign each tumor to an intrinsic molecular subtype—luminal A, luminal B, HER2 enriched, or basal like.15 Regardless of the intrinsic molecular subtype, there was a similar reduction in the risk of recurrence with the addition of abemaciclib to adjuvant endocrine therapy, as you can see here. Although the number of patients in the HER2-enriched and basal-like subsets were small, the HR values still favor abemaciclib.

There had been some concern that the basal-like subtype might not benefit from adjuvant abemaciclib based on data suggesting limited benefit with CDK4/6 inhibition in the metastatic setting among patients with hormone receptor–positive basal-like cancer.17 Fortunately, we did not see that phenomenon in this analysis of EBC.

monarchE Molecular Profiling: iDFS Across Inferred Oncotype Risk Scores

Joyce O’Shaughnessy, MD:
Using the RNAseq data, the investigators were able to infer the 21-gene Oncotype recurrence score.15 Based on this inferred Oncotype risk score, 71% of patients were categorized as having a high-risk recurrence score. Abemaciclib was associated with a comparable reduction in the risk of recurrence in patients categorized as high risk and low risk.

monarchE Molecular Profiling: iDFS Across Most Common Oncogenic Alterations

Joyce O’Shaughnessy, MD:
The investigators also explored the relationship between iDFS benefit with abemaciclib and oncogenic alterations that had a prevalence ≥9%, such as mutations in PIK3CA or TP53 and amplification of CCND1.15

Of these alterations, only MYC amplification was associated with decreased benefit from the addition of abemaciclib. This is consistent with preclinical data suggesting that c-Myc upregulation mediates CDK4/6 resistance.18 However, I would consider this finding to be hypothesis generating only and requiring further exploration and confirmation in other studies.

Technical Feasibility of ctDNA Detection in monarchE: Pilot Study Design and Patient Subset Characteristics

Joyce O’Shaughnessy, MD:
Turning now to the second biomarker analysis of monarchE, Loi and colleagues16 presented results from a pilot study on the technical feasibility of ctDNA detection and dynamics to predict recurrence in a subset of monarchE participants.

The investigators preselected 178 patients with a range of tumor mutation burden based on existing WES data from primary tumors. This subset was limited to those who had received adjuvant chemotherapy and initiated endocrine therapy before randomization and excluded patients who experienced an iDFS event within the 2-year abemaciclib treatment period.

Archival primary breast cancer tissue was collected at study entry, and plasma samples were collected at Months 0 and 24—bracketing the 2-year abemaciclib treatment period. To detect ctDNA, the investigators used the Signatera ctDNA assay, which was individualized to each patient using baseline WES data.

Technical Feasibility of ctDNA Detection in monarchE: Association Between ctDNA Positivity and Recurrence

Joyce O’Shaughnessy, MD:
After completing 2 years of adjuvant abemaciclib, 42 (23.6%) patients were ctDNA positive, and all patients experienced recurrence.16 Thus, the positive predictive value for being ctDNA positive at Month 24 was 100%.

However, 136 (76%) patients were ctDNA negative after completing 2 years of abemaciclib and, of these, 28 (21%) experienced recurrence. This yielded a negative predictive value of ctDNA negativity predicting for lack of recurrence at Month 24 of 79%.

By contrast, the positive and negative predictive values for recurrence were both lower using ctDNA status at Month 0. Many patients who were ctDNA positive at baseline became ctDNA negative over the course of 2 years of endocrine therapy with or without abemaciclib.

monarchE Molecular Profiling and ctDNA Pilot Study: Clinical Implications

Joyce O’Shaughnessy, MD:
At this time, these results do not have immediate clinical implications: We are not using ctDNA molecular residual disease assays in clinical practice, and we need to have data on the clinical utility of ctDNA for improving patient outcomes before incorporating this assay into practice. That being said, I anticipate more investigators using ctDNA positivity to identify patients at very high risk of recurrence when designing trials assessing “second-line” adjuvant therapy.

Sara A. Hurvitz, MD:
I agree completely. I find these biomarker analyses from monarchE to be fascinating, and we are currently testing whether ctDNA can be predictive of therapeutic benefit. That being said, I echo your caution that ctDNA should not be used in routine clinical practice.

Neo-N: Nivolumab Plus Neoadjuvant Chemotherapy With or Without Lead-In Nivolumab in Stage I/II TNBC

Sara A. Hurvitz, MD:
We will next discuss a trial assessing the potential benefit of “priming” with a lead-in ICI before adding neoadjuvant chemotherapy in the setting of early TNBC. The noncomparative phase II “Neo-N” (BCT1902/IBCSG 61-20 Neo-N) trial enrolled 110 patients with stage I-IIA/B TNBC and randomized them to 2 parallel treatment arms.19 The “lead-in” arm received nivolumab alone for 2 weeks, followed by nivolumab plus carboplatin/paclitaxel for 4 cycles. The “concurrent” arm started with nivolumab plus carboplatin/paclitaxel for 4 cycles followed by nivolumab alone. Of note, these were anthracycline-free regimens.

The primary endpoint was pCR, defined as ypT0/is ypN0.

Neo-N: Baseline Characteristics

Sara A. Hurvitz, MD:
A similar number of patients were randomized to each noncomparative treatment arm.19 Overall, two thirds had stage II/III disease. One third had high levels of tumor-infiltrating lymphocytes, defined as ≥30%, and approximately one half were considered PD-L1 positive, defined as ≥1% per the SP142 assay. The median Ki-67 expression was 70%. Similar data have been reported in other studies of the TNBC population.20

Neo-N: pCR

Sara A. Hurvitz, MD:
The pCR rates were fairly similar across arms, with pCR being observed in 51% (90% CI: 39%-63%) of patients in the lead-in arm and 55% (90% CI: 43%-66%) of patients in the concurrent arm.19

Of note, a high level of tumor-infiltrating lymphocytes was the sole predictor of pCR (67% vs 46%; odds ratio: 2.47) in a multivariate logistic regression analysis.

Neo-N: Clinical Implications

Sara A. Hurvitz, MD:
Although there are no immediate clinical implications of this trial assessing a neoadjuvant ICI plus an anthracycline-free chemotherapy regimen in lower-risk early TNBC, I thought these data were quite interesting. We hope to see biomarker analyses from this trial, which could help guide further investigations into ICI-based therapy for early TNBC.

Joyce O’Shaughnessy, MD:
Should we start the ICI before the neoadjuvant chemotherapy to improve pCR? According to this study, the answer is a definitive no.

In this trial, the overall pCR rate with neoadjuvant nivolumab plus carboplatin/paclitaxel was 53%. That is very similar to the pCR rate of 56% in a subgroup of patients with stage I-III TNBC and ER expression of 1% to 10% who received neoadjuvant pembrolizumab plus carboplatin and docetaxel in the NeoPACT trial.21 To put these results in context, the pCR rate was 64.8% among patients with stage II-III TNBC who received the KEYNOTE-522 regimen, which consists of pembrolizumab being added to neoadjuvant chemotherapy (carboplatin/paclitaxel followed by an anthracycline and cyclophosphamide) followed by adjuvant pembrolizumab monotherapy.3

Can we forego the anthracycline in this setting? We hope to get an answer from the ongoing phase III SCARLET trial (NCT05929768). This trial is comparing the KEYNOTE-522 regimen vs the anthracycline-free NeoPACT regimen in patients with early TNBC.

ALEXANDRA/IMpassion030: Adjuvant Chemotherapy With or Without Atezolizumab for Stage II/III TNBC

Sara A. Hurvitz, MD:
Continuing with the theme of ICI-based regimens in early TNBC, the open-label phase III ALEXANDRA/IMpassion030 trial is comparing adjuvant chemotherapy with or without atezolizumab followed by atezolizumab maintenance vs monitoring only in patients with early TNBC.22 This study addresses the important question of whether it is necessary to start ICI in the neoadjuvant phase, when an the invasive cancer is present to train the immune system, or if the ICI can be given only in the adjuvant phase.

Earlier, the phase III IMpassion031 trial demonstrated that adding atezolizumab to neoadjuvant chemotherapy in patients with early TNBC significantly improved the pCR rate vs chemotherapy alone (58% vs 41%, respectively; 1-sided P = .0044).23

ALEXANDRA/IMpassion030 randomized 2199 patients with resected stage II-III TNBC to receive induction with atezolizumab plus chemotherapy vs chemotherapy alone, followed by maintenance atezolizumab to complete 1 year, vs monitoring visits only.22 Of note, the chemotherapy included a taxane, anthracycline, and cyclophosphamide, but no platinum.

At SABCS 2023, Ignatiadis and colleagues22 presented an interim analysis of the primary endpoint, iDFS in the ITT population.

ALEXANDRA/IMpassion030: Efficacy

Sara A. Hurvitz, MD:
I was very interested to see that there was no iDFS benefit from adding atezolizumab to adjuvant chemotherapy in this setting.22 In the ITT population, the proportion of patients experiencing an iDFS event was essentially the same for each arm (11.5% with atezolizumab plus chemotherapy vs 10.2% with chemotherapy alone), yielding an HR of 1.12 (95% CI: 0.87-1.45; P = .37). Thus, futility was declared for the primary endpoint of this trial; we should note that the study design was changed midway to add a futility analysis.

No benefit in iDFS was observed among subgroups, and there was no OS benefit in the ITT population.

ALEXANDRA/IMpassion030: Safety

Sara A. Hurvitz, MD:
No new safety signals were observed in this trial, and atezolizumab did not compromise the administration of chemotherapy.22

ALEXANDRA/IMpassion030: Clinical Implications

Sara A. Hurvitz, MD:
To date, there are no data supporting the use of atezolizumab, a PD-L1 inhibitor, in patients with early TNBC who have already undergone resection of the primary cancer. There is still a question of whether a PD-1 inhibitor could be beneficial when used only in the adjuvant setting for early TNBC. The ongoing phase III SWOG S1418/BR006 trial (NCT02954874) is evaluating adjuvant pembrolizumab, and the ongoing phase II OXEL trial (NCT03487666) is evaluating adjuvant nivolumab with or without capecitabine, both in patients with TNBC with residual disease following neoadjuvant chemotherapy.

Despite questions about the lack of a platinum agent in the chemotherapy regimen, I am skeptical that the platinum-free chemotherapy is what led to futility in this trial.

Joyce O’Shaughnessy, MD:
I completely agree. This was strong evidence that using atezolizumab in just the adjuvant setting is simply not effective.

These results may be relevant regarding a clinical dilemma we might encounter: If I see a patient who would have been eligible for the KEYNOTE-522 regimen but who underwent surgery first (a situation we try to avoid), would I offer her the KEYNOTE-522 regimen as adjuvant therapy? ALEXANDRA/IMpassion030 suggests that there may be no benefit to adding an ICI to adjuvant chemotherapy. But, as you mentioned, the ICI in ALEXANDRA/IMpassion030 was the PD-L1 inhibitor atezolizumab instead of the PD-1 inhibitor pembrolizumab, and there was no platinum agent. It is difficult to know what to do in this scenario, but I would lean toward recommending the KEYNOTE-522 regimen as adjuvant therapy to patients with high-risk, stage II/III TNBC who underwent surgery first.

NRG Oncology/NSABP B-51/RTOG 1304: Adjuvant RNI in EBC Converting to Axillary Lymph Node Negativity After Neoadjuvant Chemotherapy

Joyce O’Shaughnessy, MD:
We will conclude our discussion of studies in EBC with a very important, long-awaited trial on locoregional disease control. The phase III NRG Oncology/NSABP B-51/RTOG 1304 trial enrolled 1641 patients with biopsy-proven axillary node–positive disease across hormone receptor–positive/HER2-negative, HER2-positive, and triple-negative breast cancer subtypes (clinical T1-3, N1, M0 disease).24 Patients had received standard neoadjuvant therapy and, at definitive surgery, were found to have become axillary node negative (ypN0) on sentinel lymph node biopsy (≥2 nodes excised), axillary lymph node dissection, or both. Patients could have undergone mastectomy or lumpectomy.

Patients who underwent lumpectomy received whole-breast radiation therapy with or without regional nodal irradiation (RNI). Patients who underwent mastectomy were randomized to receive chest wall radiation with RNI vs no radiation.

The primary endpoint was invasive breast cancer recurrence-free interval (IBCRFI), defined as the time from randomization to invasive local, regional, or distant recurrence, or death from breast cancer. Secondary endpoints include OS and safety.

NRG Oncology/NSABP B-51/RTOG 1304: Efficacy

Joyce O’Shaughnessy, MD:
The bottom line was that there was no significant difference in IBCRFI between treatment arms.24 The 5-year estimated IBCRFI rate was 91.8% among patients who received no RNI vs 92.7% among those who received RNI, yielding a nonsignificant HR of 0.88 (95% CI: 0.60-1.29; P = .51).

With the caveat that the 5 years of follow-up is early in the natural history of EBC, there seemed to be no difference in OS or distant recurrences. There was a very minor numerical difference in the 5-year estimated rate of isolated locoregional recurrences, which was 98.4% without RNI vs 99.3% with RNI.

NRG Oncology/NSABP B-51/RTOG 1304: Safety

Joyce O’Shaughnessy, MD:
As expected, there was less radiation-related toxicity among the patients who received no RNI.24

NRG Oncology/NSABP B-51/RTOG 1304: Clinical Implications

Joyce O’Shaughnessy, MD:
I anticipate that the application of these results will be hotly debated for individual patients at our tumor boards. The data suggest that we may be able to de-escalate radiation therapy in patients who present with more minimal node positivity and/or with ER-negative/HER2-positive disease, the latter of which is associated with a high rate of pCR.25 By contrast, we would likely not de-escalate in patients who presented with extensive nodal disease and who subsequently had no residual disease in ≥2 lymph nodes.

Because locoregional recurrences can occur beyond 5 years, we will need to see additional follow-up before we can fully understand the clinical implications of this trial.

Sara A. Hurvitz, MD:
I agree. These are very important results because we previously lacked data indicating whether nodal radiation benefits patients with disease that responds very well to neoadjuvant therapy. I found these results to actually be quite practice changing.