FDA Grants Expanded Access Program to Ulixertinib for MAPK Pathway Aberrant Cancer

By Matthew Fowler

xCures recently announced that the FDA granted its IND immediate Expanded Access Program for the ERK inhibitor ulixertinib to treat patients with MAPK pathway aberrant cancer.

The FDA granted the investigational new drug application for the ERK inhibitor ulixertinib (BVD-523) an immediate Expanded Access Program (EAP), according to an xCures press release.

The clinical stage biotechnology company, BioMed Valley Discoveries, is developing ulixertinib to treat patients with MAPK pathway aberrant cancer, including but not limited to KRAS, NRAS, HRAS, BRAF, MEK, and ERK mutations.

“xCures prospective real-world evidence generation capability transforms managed access programs such as the ulixertinib expanded access program by making them an efficient way for physicians and patients to gain access to promising therapies when clinical trials are not an option,” Mika Newton, CEO of xCures, Inc., said in a press release.

The Expanded Access Program provides ulixertinib for compassionate use in patients with advanced cancer and a MAPK pathway-altered solid tumor(s). These patients have also exhausted all previously available lines of therapy.

The program collects data about a patient’s treatment to provide a complete and accurate case report to health authorities. These authorities use the data to “assess response to treatment, safety, tolerability, and quality-of-life.”

The Expanded Access Program involves the use of an unapproved drug for the treatment of patients with serious or life-threatening illnesses outside of a clinical trial setting.

“xCures’ programs uniquely capture high-value evidence related to the safety and efficacy from this expanded set of patients,” said Newton in a press release.

The best-in-class small-molecule inhibitor of extracellular signal-regulated kinase (ERK) family kinases (ERK1 and ERK2), ulixertinib, has demonstrated promising early efficacy for patients with “tumors harboring alterations in the MAPK pathway.”

According to xCures, this EAP is open across the United States to adolescent and adult patients with cancer who cannot access an open clinical trial to investigate ulixertinib.

Reference:

xCures announces the launch of a Compassionate Use program for ulixertinib (BVD-523) [news release]. San Francisco, California. Published September 28, 2020. https://www.prnewswire.com/news-releases/xcures-announces-the-launch-of-a-compassionate-use-program-for-ulixertinib-bvd-523-301139076.html. Accessed September 29, 2020.

This article was published by Cancer Network.

Bayer’s ATR Inhibitor May Benefit Cancers With ATM Aberrations, Early Data Suggest

Researchers are optimistic that data from a first-in-human clinical trial of Bayer’s ATR-inhibiting agent, BAY-1895344, may pave the way for a new class of precision oncology treatments for patients with certain DNA damage response (DDR) defects in their tumors.

The study results, published Tuesday in the journal Cancer Discovery, suggest that patients whose tumors have ATM protein loss or deleterious ATM mutations may be particularly sensitive to ATR inhibition.

The Phase I clinical trial of BAY-1895344 includes patients with advanced solid tumors or lymphomas, though the recently published paper detailed the results from 21 patients with solid tumors, including those with breast, colorectal, prostate, ovarian, and endometrial cancers, among others. The majority of patients had received more than four prior lines of chemotherapy.

Half of the enrolled patients had one or more ATM aberrations detected by next-generation sequencing or immunohistochemistry analysis of baseline tumor biopsies. Six patients had both an ATM deleterious mutation and loss of ATM protein expression; two patients had ATM deleterious mutations with normal ATM protein expression; and three patients had loss of ATM protein expression and no ATM mutations. Four patients had normal ATM protein expression and no mutations in the gene. Beyond ATM, three patients’ tumors had BRCA1 mutations and one had a BRCA2 mutation.

As a first-in-human trial, the primary aim of the study was to evaluate the safety and maximum tolerated dose of BAY-1895344, though anti-tumor activity was assessed as a secondary endpoint. The objective response rate was 19 percent among all patients evaluated and 36.4 percent among those with ATM loss and/or ATM mutations.

In terms of safety, the investigators, led by Johann de Bono of the Institute of Cancer Research in London and Timothy Yap of MD Anderson Cancer Center in Texas, noted that the drug was tolerated well when dosed in an intermittent schedule of three days on followed by four days off. ATR is essential for the functioning of normal cells, and the days off treatment, researchers noted, allowed for the recovery of normal tissue.

Most of the adverse events observed, such as anemia, were hematological in nature, leading the authors to conclude that future studies attempting to combine ATR inhibition with chemotherapy should be approached with caution, since systemic chemo agents are also known to cause hematological toxicities.

The authors of the Cancer Discovery paper highlighted that one patient with a deleterious BRCA1 mutation experienced stable disease and a partial response for over a year on the ATR inhibitor. This case was particularly noteworthy because the patient had previously progressed after treatment with chemotherapy, immunotherapy, and the PARP inhibitor olaparib (AstraZeneca/Merck’s Lynparza).

“The clinical benefit observed in this patient following BAY-1895344 monotherapy is of particular interest in view of preclinical data suggesting that acquired PARP inhibitor resistance may be mediated by ATR-induced protection of the replication fork and is a clinical area of unmet need,” wrote the authors. In other words, ATR inhibitors may potentially be coupled with PARP inhibitors.

Indeed, Bayer has an additional ongoing Phase Ib clinical trial exploring the combination of BAY-1895344 with the PARP inhibitor niraparib (GlaxoSmithKline’s Zejula) as a treatment for patients with ovarian cancers and other advanced solid tumors. Evidence of up-regulation of PD-L1 expression after treatment with BAY-1895344 in patients whose tumors were PD-L1 positive also bolstered the case for another advanced solid tumor Phase I clinical trial, in which Bayer is studying the ATR inhibitor in combination with the PD-L1 inhibitor pembrolizumab (Merck’s Keytruda).

According to the authors, the study of BAY-1895344 is, to their knowledge, the first study to provide clinical evidence of an oral ATR inhibitor with durable single-agent antitumor activity in patients with advanced cancers with ATM aberrations.

The investigational drug is now being evaluated in a dose expansion phase of the study, specifically as a treatment for patients with DDR deficiencies resulting from genetic mutations or the loss of ATM protein expression as determined by IHC.

“It is very promising to see patients responding in an early-stage trial like this,” wrote de Bono in a statement about the results, adding that ATR inhibition with BAY-1895433 may become a new form of targeted treatment. “We are looking forward to further clinical trials to test the drug’s efficacy.”

This article was published by Precision Oncology News.

Halting Cancer by Halting DNA Repair

By Liam Drew

PARP inhibitors are rapidly transforming the treatment of ovarian, breast, prostate and other types of cancer. To develop these drugs, researchers supported by Cancer Research UK had to decipher how blocking DNA repair could expose a weak point in the biology of cancer cells.

In the early 1990s, Steve Jackson seemed to have his career all planned out. Freshly recruited to the University of Cambridge as a group leader at the Gurdon Institute, he was going to keep doing what he’d done so successfully as a postdoc—study the fundamentals of gene transcription. But then came an experiment that changed everything.

His lab’s first graduate student, Tanya Gottlieb, was given the task of investigating the function of an enzyme Jackson had “stumbled across” during his postdoc. This enzyme was activated when it bound to DNA, but Gottlieb and Jackson discovered that it wasn’t activated by DNA alone—it was stimulated by DNA breakages.

The finding swept Jackson into the emerging field of DNA repair, and, from there, into cancer research.

Jackson soon hit upon an idea—could inhibiting DNA repair enzymes aid in killing cancerous cells? The thinking is counterintuitive. Within the cells of a human body, DNA is constantly breaking—remarkably, the genome of each cell sustains around 100,000 ruptures a day. By restoring the integrity of broken DNA—and, so, preventing mutations—the body’s DNA repair mechanisms protect us from cancer. Blocking repair would, therefore, seem to be a highly dangerous thing to do.

However, Jackson drew on the fact that chemo- and radiotherapy are partly predicated on the increased susceptibility of rapidly dividing cancer cells to DNA damage. Moreover, such therapies massively activate DNA repair systems, and the cancer cells that survive such treatments rely heavily on these systems. It seemed plausible, therefore, that suppressing DNA repair might make existing cancer therapies more effective.

Two hundred miles north of Cambridge, researchers at Newcastle University were working on the same concept. They were focusing specifically on a previously discovered DNA repair enzyme called PARP. This protein binds to, and is activated by, single-stranded breaks in DNA, synthesizing a chain of ADP-ribose molecules that recruit the enzymatic machinery that will repair that breakage. Based on a growing body of academic work, the Newcastle team believed that a PARP inhibitor administered alongside either chemotherapy or ionizing radiation would help destroy tumors.

Soon, Jackson’s attention also turned to PARP and, in parallel, these two research groups pushed forward drug discovery programs that led to the clinical approval of PARP inhibitors for cancer treatment.

The drug born of Jackson’s research is olaparib, which is marketed as Lynparza by AstraZeneca. The Newcastle team developed rucaparib, initially developed by Agouron Pharmaceuticals and now sold as Rubraca by Clovis Oncology. After the promise of these two pioneering medicines became apparent, two other drugs—niraparib and talazoparib—were developed by other pharmaceutical companies.

To get here, however, the Cambridge and Newcastle groups had to overcome a problem: no matter how appealing their idea was in theory, pharmaceutical companies were too wary of the dangers of interfering with DNA repair to invest heavily in this approach.

Each group succeeded through partnering with The Cancer Research Campaign—one of the two charities that merged to form Cancer Research UK (CRUK) in 2002. The charity was willing to invest in the type of risky, blue skies research that makes big pharma nervous.

Independently, the two groups made a vital discovery that led them to fully realize the potential of PARP inhibitors. In collaboration with other academic groups (including Alan Ashworth’s team at the Institute of Cancer Research in London, which is part-funded by CRUK), they found that PARP inhibitors can target a particular weakness of cancers driven by mutations in BRCA genes.

Currently, PARP inhibitors are approved for treating BRCA-mutation-associated ovarian, breast, Fallopian tube, pancreatic and prostate cancers as second-line and, increasingly, as first-line treatments. It is also becoming increasingly clear that olaparib and rucaparib are effective in treating certain forms of BRCA-mutation-negative cancers, and research is ongoing to further explore such promising opportunities.

Today, over 30,000 patients have been treated with olaparib and that number is growing rapidly. Jackson is therefore able to say that his basic research—and his drive to commercialize it—has saved and extended people’s lives. Looking back, he says of his relationship with the Newcastle group that “some friendly competition is a good thing—you know you’re in an exciting place when there’s competition.”

“But,” he adds purposefully, “this isn’t about two antagonistic groups. It’s all about academic groups translating their science to make the world a better place.”

Newcastle and the idea of a sensitizer

Ruth Plummer is a consultant medical oncologist at Newcastle University. In 2003, she wrote the first ever prescription for a patient to receive a drug designed to suppress DNA repair.

That moment came eight years after Plummer had joined the Newcastle team and got excited by the promise of their nascent PARP inhibitor project. It was an appointment that marked a turnaround in her career. Plummer had done a Ph.D. in neuroscience while completing her medical degree, but she clicked with cancer specialist Hilary Calvert, the head of Newcastle’s clinical trials unit, and she felt strongly that “in oncology, things were changing—or had the potential to change.”

Research into PARP at Newcastle, Plummer says, was inspired largely by a 1980 paper showing that cells in a dish treated with a DNA-damaging agent died in greater numbers if PARP was simultaneously inhibited than in the absence of PARP inhibition.

The compound used in that paper was a weak inhibitor of PARP with no prospects of becoming a clinically useful drug. But it was good enough to conduct experiments that supported the hypothesis that more cancer cells died if PARP was inhibited during chemo- or radiotherapy.

The Newcastle team looked to The Cancer Research Campaign for support and together they searched around to see if anyone was developing drugs that blocked PARP. They weren’t.

Therefore, The Cancer Research Campaign funded Newcastle’s initial work and then financed the creation of a Drug Discovery Unit there. That unit was led by Herbie Newell and Calvert, and included an in-house medicinal chemistry team, headed by Bernard Golding and Roger Griffin, which developed a panel of promising PARP inhibitors.

The emergence of these compounds led to a partnership with Agouron Pharmaceuticals, a small San Diego biotech startup specializing in early drug development. In 1999, as Plummer and colleagues pushed toward a first test of these drug in patients, a subsidiary of Pfizer bought Agouron Pharmaceuticals. The Cancer Research Campaign commercial team had a crucial role in this acquisition and in shepherding the drugs toward clinical testing. They led negotiations and after the deal they continued to work with Pfizer in various ways, including the management of patents.

Despite these investments from Pfizer, however, there were still cold feet about the drug’s safety when time came to run the first PARP inhibitor clinical trial. So, the Center for Drug Development at the newly forged CRUK—established to take risky projects through early phase clinical research—stepped in to sponsor the trial.

This trial, which began in 2003, tested rucaparib in combination with a chemotherapeutic agent in patients with various tumors including late-stage melanoma. The hope was that inhibiting PARP would heighten the effects of chemotherapy. But, as with any Phase 1 trial, the priority was to determine if rucaparib was safe and tolerated by patients. By examining biopsies of the patients’ tumors, Plummer also wanted to confirm that the drug was able to get into human tumors and inhibit PARP there.

“We were very cautious when we went into the clinic,” Plummer says. “We thought it would be alright, but nobody had done it… We built a big safety factor into the trial.” Dosing started low and she was also open and clear with the patients about both the risks involved and the wider goals. “I did what I do in clinical practice all the time,” she says. “I told the patients, ‘We hope we might have a new and better drug, but we don’t know.'”

The study showed that the drug was, indeed, safe and—as Plummer puts it—”that it does what it says on the tin.” The question was now how best to use it.

Frustratingly, it became apparent that although a PARP inhibitor was fairly safe when used alone, it was difficult to use in combination with another chemotherapy agent, as together the two drugs caused serious side-effects at doses below those needed for cancer treatment. Furthermore, in this first trial, the gains in slowing tumor growth were inconsistent and only weak to moderate, and overall survival did not improve.

However, while this clinical work proceeded, two pivotal meetings—one in a seminar room, one in a bar—changed everything.

BRCA to the Future

In the early 2000s, at a conference in Oxford, Jackson had a late-night conversation with Ashworth, who was then working at the Institute of Cancer Research in London. Jackson had by then helped develop an effective PARP inhibitor. Ashworth was an expert on BRCA-mutation-associated cancers. Their chat led to a groundbreaking experiment.

Jackson had sought an industrial partner to explore his idea that blocking repair enzymes would help treat cancer.

However, as had happened with the Newcastle team, the big drug companies Jackson spoke to weren’t interested; the risk was perceived as too great. So, with the support of the University of Cambridge, Jackson turned to The Cancer Research Campaign to help him spin out his own company.

In addition to working with the university’s Technical Transfer Office to file patents, the charity provided seed funding and support to Jackson, and in December 1997 KuDOS Pharmaceuticals was born. The founders then worked together to secure significant venture capital funding, so that by mid-1999 the company was up and running.

Using the Jackson lab’s biological expertise and the various assays it had developed, KuDOS quickly identified promising molecules for inhibiting PARP and other repair enzymes. Jackson says KuDOS’s early reasoning was similar to Newcastle’s—that PARP inhibition might sensitize cancer cells to existing DNA-targeting therapies. However, his thinking was also strongly shaped by the concept of synthetic lethality.

As Jackson explains, many fundamental cellular functions are achieved by two or more pathways working towards essentially the same endpoint. Such functional overlap makes biological systems robust to genetic or drug-induced disruption of a single pathway. This means that disrupting one of the functionally overlapping pathways often has no obvious effect on a cell. But if all the pathways are halted, the cell dies.

Jackson speculated that this principle would apply to drugs disrupting the vital task of DNA repair, which involves multiple partially overlapping pathways. After all, PARP inhibitors were relatively safe—evidence that inhibiting this specific DNA repair pathway alone didn’t have overtly deleterious effects on human cells. But what if two pathways were disrupted?

That night in Oxford, Ashworth told Jackson about his work on BRCA mutations. Inheriting a faulty copy of either BRCA1 or BRCA2 drastically increases a person’s risk of developing cancer, particularly breast or ovarian—although disease only occurs when cells undergo mutations that compromise or delete the patient’s remaining functional copy of BRCA1 or BRCA2.

BRCA1 and BRCA2 encode DNA repair proteins that help repair double-stranded breaks. Jackson and Ashworth speculated that cells lacking a functional BRCA-dependent repair pathway might be relying more heavily on PARP-mediated DNA repair to survive. These cells might therefore be killed by a drug that took out this second pathway.

The two scientists realized they had perfectly complementary resources: Ashworth had cell lines lacking BRCA genes and Jackson had PARP inhibitors. A powerful collaboration was immediately struck, and straight after the conference Jackson and his colleagues at KuDOS had the drugs sent to Ashworth’s lab.

Plummer recalls a near identical exchange at Newcastle. Thomas Helleday, who was then at Sheffield University, visited Newcastle to give a seminar about his work on BRCA genes and used essentially the same logic to lay out the case for why cells lacking BRCA1 or BRCA2 might be killed by a PARP inhibitor. Nicola Curtin, the leader of Newcastle’s drug development biology program, immediately offered Helleday the drugs he needed to test the idea.

The two sets of collaborators were aware of each other’s research but worked independently. First using cultured cells, then using tumors lacking BRCA genes grafted into mice, both groups found that their PARP inhibitor given at certain doses killed cells without a BRCA gene, whereas cells containing either one or two copies of that BRCA gene survived.

These research findings suggested that PARP inhibitors—at least for certain patients with certain cancers—might achieve that goal. The two groups agreed to submit their work nearly simultaneously and their papers came out back-to-back in the 14 April 2005 issue of Nature.

Into the clinic

“Within six weeks of the papers coming out we’d gone back to CRUK,” Plummer says, “requesting support to test rucaparib in patients with BRCA mutations.”

The trial was quickly agreed, although some delays meant it didn’t commence until 2007. Excitingly, Plummer saw the tumor of the second patient she treated shrink.

KuDOS’s Phase 1 clinical trial of olaparib started in mid-2005 and then was taken forth by AstraZeneca, which acquired KuDOS in December that year. That trial involved 60 patients with late-stage breast, ovarian or prostate cancer—22 of whom were carriers of BRCA1 or BRCA2 mutations. The drug was found to be safe in all patients. Much more strikingly, olaparib clearly reduced tumor size in around half of the BRCA mutation carriers—especially in people with ovarian cancer.

Jackson recalls vividly KuDOS’s development director, Peter Harris, announcing at a board meeting that one of the first treated patients had a dramatic reduction in tumor size, calling it “a real eureka moment.”

Subsequent work homed in on BRCA mutation carriers and on ovarian cancer, with Phase 2 and 3 trials confirming the effectiveness of olaparib and rucaparib against these tumors. In December 2014, the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) approved olaparib for treating ovarian cancer in carriers of BRCA1 or BRCA2 mutations who’ve previously undergone chemotherapy. Two years later, the FDA gave rucaparib accelerated approval for similar indications.

Other pharmaceutical companies developed different PARP inhibitors, two of which have now been FDA-approved. Zejula (niraparib), developed by Tesaro, was approved for ovarian and Fallopian tube cancers in 2017. Talzenna (talazoparib), developed by Pfizer, was approved in 2018 for the treatment of patients with deleterious or suspected deleterious germline BRCA-mutated HER2-mutation-negative locally advanced or metastatic breast cancer.

In December 2019 and June 2020, respectively, the FDA and EMA approved olaparib for BRCA1- and BRCA2-mutated metastatic pancreatic cancers, and in May 2020 the FDA approved olaparib for BRCA1- and BRCA2-deficient metastatic prostate cancers.

Jackson is excited by the use of PARP inhibitors as first-line treatments. In his office, he brings up on screen the Kaplan-Meier curves from three successive trials of olaparib for BRCA-mutation-associated ovarian cancer. As the drug was given earlier and earlier to patients, the further apart the survival curves of the drug and placebo groups became. Pointing to the third one, he beams, “that includes cures!”

Plummer says she too has several patients she treated with PARP inhibitors who have gone into complete remission. “Not many,” she says, “but it can happen and that is fantastic.”

Where to now?

As clinicians increasingly refine the use of PARP inhibitors in patients with inherited BRCA mutations, there is also a drive to identify people who don’t carry germline BRCA mutations but whose cancers might also be sensitive to PARP inhibitors. These include tumors with somatic loss of BRCA gene function and tumors that don’t lack BRCA function but which have deficits in this DNA repair pathway via some other means—a concept termed “BRCAness.”

An important research focus now is therefore to develop better biomarkers and other tests for identifying tumors that will be sensitive to PARP inhibitors. And that includes discovering signals that might say if a cancer from a BRCA1 or BRCA2 mutation carrier will be resistant to this drug class.

Plummer is also currently running trials to test whether combining a PARP inhibitor with cancer immunotherapy is useful. These were inspired by work from other groups showing that blocking PARP can increase the immunogenicity of tumor cells and, so, enhance the effectiveness of the rapidly evolving immunotherapy approach to cancer.

Jackson remains focused on discovery science, while forever seeking ways in which his work might inform clinical medicine. Among numerous projects in his lab, he’s particularly interested in the mechanisms of resistance to PARP inhibitors, and how they might be overcome. “Resistance to one drug can give you sensitivity to another drug,” he says. That potential second drug might target another aspect of DNA repair, he says, noting that two of his old colleagues at KuDOS, Niall Martin and Graeme Smith, now lead a company called Artios Pharma, which is developing such drugs. “But that’s all for the future,” he says.

This article was published by Medical Xpress.

Immunotherapy Falls Short Again in Ovarian Cancer

No PFS benefit in advanced disease with addition of atezolizumab to chemo, bevacizumab.

By Charles Bankhead

An immunotherapy boost failed to improve outcomes in newly diagnosed advanced ovarian cancer compared with chemotherapy and bevacizumab (Avastin), a large international trial showed.

Progression-free survival (PFS) in the overall study population increased about a month with the addition of atezolizumab (Tecentriq) to carboplatin-paclitaxel chemotherapy and bevacizumab. Median PFS in the PD-L1-positive subgroup (≥1% expression) improved by 2.3 months (20.8 vs 18.5 months), but the difference did not meet prespecified criteria for statistical significance. An exploratory analysis suggested improved PFS with atezolizumab in the subgroup of patients with PD-L1 immune cell (IC) expression ≥5%.

A preliminary analysis of overall survival (OS) also suggested no benefit from the addition of atezolizumab to the standard regimen, Kathleen Moore, MD, of the University of Oklahoma Sciences Center in Oklahoma City, reported at the 2020 European Society for Medical Oncology (ESMO) virtual congress.

“Despite notable successes with incorporation of atezolizumab and bevacizumab in the treatment of other solid tumors, IMagyn50, the first such study in ovarian cancer, did not meet its first primary endpoint for extending progression-free survival in either the intention-to-treat or fully powered PD-L1-positive population,” said Moore. “The signal of a clinical benefit for atezolizumab in the immune cell PD-L1-high subgroup may warrant further evaluation. The interim analysis of overall survival showed no differences between treatment groups, but a final analysis will be expected and presented in 2023.”

The findings had a precedent in a trial reported earlier this year at a Society of Gynecologic Oncology webinar. A phase III trial of chemotherapy plus avelumab (Bavencio) not only failed to improve PFS versus chemotherapy alone but suggested a possible harm with the addition of anti-PD-1/L1 therapy. That trial was preceded by a negative randomized phase III study comparing pegylated liposomal doxorubicin (PLD) and avelumab versus PLD alone in patients with advanced platinum-resistant/refractory ovarian cancer.

Background and Trial Design

Atezolizumab has demonstrated efficacy in multiple solid tumors. Platinum-taxane chemotherapy is an established first-line regimen for advanced ovarian cancer, and combining angiogenesis inhibition with PD-1/L1 blockade has demonstrated antitumor efficacy in several types of advanced solid tumors, Moore noted. Collectively, the evidence supported a trial to evaluate the addition of atezolizumab to chemotherapy and bevacizumab.

The IMagyn050 trial involved patients with untreated stage III/IV epithelial ovarian, primary peritoneal, or fallopian tube cancer. Patients had postoperative macroscopic residual disease or planned neoadjuvant chemotherapy with planned interval surgery. All patients received carboplatin, paclitaxel, and bevacizumab and were randomized to placebo or atezolizumab.

The trial had co-primary endpoints: PFS in both the PD-L1-positive and intention-to-treat (ITT) populations (simultaneously tested) and OS in the same two populations, with hierarchical testing.

Data analysis included 1,301 patients, who had a median age of 59-60. A fourth of the patients had received neoadjuvant therapy and the rest underwent primary surgery. Moore said 60% of patients had PD-L1-positive tumors, and 69% had stage III disease.

Key Findings

The results showed a median PFS in the ITT population of 19.5 months with atezolizumab and 18.4 months without it. The slightly larger difference in the PD-L1-positive group was associated with conventional statistical significance (P=0.0376) but the point estimates for the two groups overlapped.

Median OS had yet to be reached for either group in the ITT or PD-L1-positive analyses. The 2-year event-free survival rate was about 80% for each group in the ITT analysis and about 82% for the PD-L1-positive analysis. A subgroup analysis did not identify any patients who appeared to benefit more with the addition of atezolizumab. An analysis of PFS by histology also failed to show a significant advantage for atezolizumab.

Investigators performed an exploratory analysis of PFS by the proportion of PD-L1-positive ICs and the presence or absence of tumor cell (TC) staining. The results yielded evidence of improved PFS with atezolizumab in the subgroup with IC ≥5%. Placebo-treated patients had a median PFS of 20.2 months, whereas the median had yet to be reached in the atezolizumab arm. The difference represented a 36% reduction in the hazard for progression or death (95% CI 0.43-0.96, P=0.0278). The subgroup accounted for 20% of the total study population, said Moore.

The analysis of TC staining also suggested a PFS benefit with atezolizumab (15.0 months vs not evaluable, HR 0.41, 95% CI 0.19-0.90), but the group represented only 5% of the study population, she added.

The safety profile was similar between the two groups, with a few exceptions, although atezolizumab was associated with more serious adverse events (47% vs 33% overall and 35% vs 21% considered treatment related) and more adverse events leading to discontinuation of atezolizumab versus placebo (15% vs 6%).

Despite the negative results from IMagyn050 and the avelumab studies, the death knell might not have sounded for PD-1/L1 in ovarian cancer, said ESMO-invited discussant Isabelle Ray-Coquard, MD, PhD, of Claude Bernard University in Lyon, France. Longer follow-up is needed in IMagyn050 for both PFS and OS. In the meantime, the first priority should be to learn more about the subgroup with PD-L1 IC expression ≥5% (BRCA and homologous recombination deficiency status, histology, surgical outcome).

Evaluation of immunotherapy should move beyond high-grade serous ovarian cancer to the more uncommon subtypes, Ray-Coquard continued. Different sequences of chemotherapy and immunotherapy require exploration, as well as the impact of integrating a PARP inhibitor. Finally, alternative chemotherapy backbones should be explored in the setting of relapsed platinum-sensitive disease.

This article was published by MedPage Today.

Why Some Cancers May Respond Poorly to Key Drugs Discovered

Patients with BRCA1/2 mutations are at higher risk for breast, ovarian and prostate cancers that can be aggressive when they develop—and, in many cases, resistant to lifesaving drugs. Now scientists at The University of Texas at Austin and Ajou University in South Korea have identified a driver of the drug resistance that can make a life or death difference for patients with these cancers.

“A major issue with cancer treatments is the development of resistance,” said Kyle Miller, a UT Austin associate professor of molecular biosciences. “When treatments stop working for patients, it’s incredibly demoralizing and it’s been a huge drive in research to understand these resistance mechanisms.”

In a paper published today in the journal Molecular Cell, the researchers describe a protein that may help doctors predict which patients will become resistant to a class of drugs frequently used to treat BRCA 1/2-deficient tumors. The finding could help create more effective treatment plans for their patients.

The scientists identified that a protein called PCAF promotes DNA damage in BRCA 1/2-mutated cancer cells. Patients with low levels of this protein are likely to have poor outcomes and develop resistance to a type of drug that is used to treat BRCA-deficient tumors, called a PARP inhibitor.

“PARP inhibitors are an important breakthrough in treating these aggressive cancers,” Miller said. “What we found is that when levels of PCAF are low, it actually protects the cancer cells from this drug. By testing biopsy samples, doctors may be able to tell using PCAF as a molecular marker for PARP inhibitor responses what treatment may work best for a patient.”

Fortunately, there is already another class of drugs on the market, called HDAC inhibitors, that can boost the effectiveness of the PCAF protein. HDAC inhibitors and PARP inhibitors have the potential to be prescribed as a combination therapy.

“Previous studies have shown that these two drugs work well together,” Miller said. “We believe we’ve found the reason why.”

It is possible to test for PCAF levels in biopsy or tissue samples, Miller said, and in the future, the test could be included on a standard panel for cancer testing.

But unlocking the workings of PCAF doesn’t just offer clues to combatting cancer. Because this protein is responsible for modifying chromatin, the stuff that organizes 6 feet of DNA in each of our cells so that it fits into its nuclear volume, PCAF also may offer important clues about cell replication.

“The focus in my lab is on understanding chromatin and its impact on replicating DNA, protecting DNA and controlling access to DNA,” Miller said. “Our goal is to understand how every molecule is interacting inside our cells, as this gives clues to what is going wrong in human diseases.”

This article was published by Medical Xpress.

Olaparib Improves Median Time Without Disease Progression in BRCAm Ovarian Cancer

Olaparib (Lynparza®; AstraZeneca/Merck & Co.*) has demonstrated a long-term progression-free survival (PFS) benefit versus placebo as a 1st-line maintenance treatment in patients with newly diagnosed, advanced BRCA-mutated (BRCAm) ovarian cancer who had a complete or partial response following platinum-based chemotherapy.

Ovarian cancer, which includes ovarian, fallopian tube, and primary peritoneal cancers,  ranks fifth in cancer deaths among women in the U.S., and in 2020, an estimated 21,750 women in the United States will receive a new diagnosis and about 13,940 women will die from ovarian cancer. It is the fifth leading cause of cancer death among women in the U.S.

The risk of developing ovarian cancer is increased in women with specific inherited genetic abnormalities, including BRCA mutations.

PARP inhibitor
Olaparib a first-in-class PARP inhibitor and the first targeted treatment to block DNA damage response (DDR) in cells/tumors harboring a deficiency in homologous recombination repair, such as mutations in BRCA1 and/or BRCA2.

Poly (ADP-ribose) polymerase (PARP) is a family of proteins involved in a number of cellular processes such as DNA repair, genomic stability, and programmed cell death. Inhibition of with olaparib leads to the trapping of PARP bound to DNA single-strand breaks, stalling of replication forks, their collapse, and the generation of DNA double-strand breaks and cancer cell death. The drug is being tested in a range of PARP-dependent tumor types with defects and dependencies in the DDR pathway.

SOLO-1 trial
SOLO-1 was a Phase III randomized, double-blinded, placebo-controlled, multi-center trial to evaluate the efficacy and safety of olaparibtablets (300 mg twice daily) as a maintenance monotherapy compared with placebo in patients with newly diagnosed BRCAm advanced ovarian cancer following first-line platinum-based chemotherapy. The trial randomized 391 patients with a deleterious or suspected deleterious germline or somatic BRCA1 or BRCA2 mutation who were in clinical complete or partial response following platinum-based chemotherapy.

Patients were randomized (2:1) to receive olaparibor placebo for up to two years or until disease progression. Patients who had a partial response at two years were permitted to stay on therapy at the investigator’s discretion. The primary endpoint was PFS and key secondary endpoints included time to second disease progression or death, time to first subsequent treatment, and overall survival.

Five-year follow-up data from the Phase III SOLO-1 trial showed olaparib reduced the risk of disease progression or death by 67% (based on a hazard ratio [HR] of 0.33; 95% confidence interval [CI] 0.25-0.43) and improved PFS to a median of 56.0 months versus 13.8 months for placebo. At five years, 48.3% of patients treated with olaparib remained free from disease progression versus 20.5% on placebo. The median duration of treatment with olaparib was 24.6 months versus 13.9 months with placebo.

“For patients with newly diagnosed BRCA-mutated advanced ovarian cancer, the benefit derived from two years of maintenance treatment with olaparib continued long after treatment ended. After five years, almost half of these women were free of cancer progression,” said Susana Banerjee, MBBS MA Ph.D., FRCP, one of the investigators from the SOLO-1 trial and Consultant Medical Oncologist at The Royal Marsden NHS Foundation Trust and Reader at the Institute of Cancer Research, London.©

“These results represent a significant step forward in the treatment of BRCA-mutated ovarian cancer,” Banerjee added.

“Once a patient’s ovarian cancer recurs, it has historically been incurable. Even at an advanced stage, we have shown that maintenance treatment with olaparib can help patients achieve sustained remission,” José Baselga, M.D., Ph.D., Executive Vice President, Oncology R&D at AstraZeneca.

“Today’s results further underline the critical importance of identifying a patient’s biomarker status at the time of diagnosis to be able to offer a treatment that may help delay disease progression,” Baselga noted.

“This is the first trial of a PARP inhibitor to read out a five year follow up and showed olaparib improved progression-free survival to over four and a half years versus 13.8 months with placebo following response to 1st-line platinum-based chemotherapy. This latest data represents a major and significant milestone in a disease that has historically had such a poor prognosis,” said Roy Baynes, M.D., Ph.D., Senior Vice President and Head of Global Clinical Development, Chief Medical Officer, Merck & Co (MSD).

The safety profile of olaparib was consistent with previous observations. The most common treatment-related adverse events (TRAEs) ≥20% were nausea (77%), fatigue/asthenia (63%), vomiting (40%), anemia (39%) and diarrhea (34%). The most common ≥ grade 3 AEs were anemia (22%) and neutropenia (9%). Twelve percent of patients on olaparib discontinued treatment due to a treatment released adverse event.

The results were presented on Friday, September 18, during the 2020 European Society of Medical Oncology (ESMO) virtual congress.

The Phase III SOLO-1 trial met the primary endpoint of PFS in June 2018, which formed the basis of approvals in the US, the EU, Japan, China, and several other countries.

Note
* In July 2017, AstraZeneca and Merck & Co. (MSD) entered a global strategic oncology collaboration to co-develop and co-commercialize olaparib.

Clinical trials
Olaparib Maintenance Monotherapy in Patients With BRCA Mutated Ovarian Cancer Following First Line Platinum-based Chemotherapy. (SOLO-1) – NCT01844986

This article was published by Onco’Zine.

Phase I Data of XMT-1536 Supports Continued Development in Ovarian Cancer

Updated interim safety, tolerability, and efficacy data for the ovarian cancer cohort of the ongoing expansion portion of the Phase I study evaluating XMT-1536 show a 34% objective response rate (ORR) and 79% disease control rate, including two complete responses. The investigational antibody-drug drug, being developed by Mersana Therapeutics, targets NaPi2b, a multi-transmembrane sodium-dependent phosphate transporter expressed in human ovarian cancers.

The NaPi2b antigen is broadly expressed in ovarian cancer and non-small cell lung cancer (NSCLC) adenocarcinoma. The investigational agent is produced via Mersana’s proprietary Dolaflexin platform to deliver an average of 10-12 DolaLock payload molecules per antibody.

The DolaLock payload, a  proprietary auristatin cytotoxic drug, controls the bystander effect by locking the cytotoxic drug inside cells after allowing a short period of diffusion throughout the tumor. As the drug diffuses through neighboring tissue, the DolaLock payload is metabolized to a form that is still highly potent but no longer able to cross the cell membrane. DolaLock effectively locks the drug inside cells, controlling the bystander effect for safer and more effective cancer therapy.

The interim analysis focused on the ovarian cancer cohort of the Phase I expansion study, including heavily pre-treated patients with platinum-resistant or refractory ovarian cancer, fallopian tube, or primary peritoneal cancer who have received up to three lines of prior therapy, and received, in some cases, four lines of prior therapy regardless of platinum status. With a data cutoff of August 18, 2020 these data included 47 patients.

The data includes an additional follow up on the 27 ovarian cancer patients previously presented at the American Society of Clinical Oncology (ASCO) virtual program in May of 2020 as well as 20 new patients who entered the study between May 1, 2020 and August 18, 2020.

Safety profile
According to the investigators, the safety profile of XMT-1536 was consistent with previously reported expansion data. The drug also continues to be generally well-tolerated and no new safety signals were observed. The most frequently reported treatment-related adverse events (TRAEs) were generally Grade 1-2 fatigue, nausea, decreased appetite, vomiting, and transient AST elevation without associated changes in bilirubin or cases of Hy’s law. There were no reported cases of severe neutropenia, peripheral neuropathy, or ocular toxicity.

Other key findings from the interim study included a continued, significant anti-tumor activity in platinum-resistant and platinum-refractory ovarian cancer and in ovarian cancer previously treated with bevacizumab, PARP inhibitors, or both. Of the 29 patients that were evaluable for response, 2/29 (7%) achieved confirmed complete responses (CRs) and 8/29 (28%) achieved confirmed partial responses (PRs) for an objective response rate (ORR) of 34%. Additionally, 13/29 (45%) patients achieved stable disease (SD). The disease control rate (DCR) was 23/29 (79%).

Durability Data are Immature: 50% of Ovarian Cancer Patients with Higher NaPi2b are Still Ongoing.

The investigators also found that 70% of responses were observed within two cycles and 100% of responses were observed within four cycles. Responses appeared to deepen over time, including responses in patients receiving reduced dose levels. The majority of responders had prior treatment with bevacizumab, PARP inhibitors, or both. Both patients with confirmed CRs had prior treatment with bevacizumab and PARP inhibitors. Reduction in tumor volume was observed in the majority of patients achieving a best response of stable disease.

Data continue to support a NaPi2b biomarker-based patient selection strategy based on depth, time on study, and quality of response. Further, 50% of patients with higher NaPi2b expression are ongoing in the study while only 33% of patients with lower Napi2b expression are ongoing in the study. The median duration of response was not yet reached in the 7 patients with ovarian cancer with higher NaPi2b expression.

The updated data was presented during the 2020 European Society of Medical Oncology (ESMO) Virtual Congress.

“These data further support the continued development of XMT-1536, our first-in-class Dolaflexin ADC targeting NaPi2b, which has recently been granted U.S. Food and Drug Administration (FDA) Fast Track Designation,” noted Anna Protopapas, President and Chief Executive Officer of Mersana Therapeutics.

Based on the updated interim results, experts believe that XMT-1536 could change the treatment paradigm for ovarian cancer and non-small cell lung cancer (NSCLC).

“We are eager to advance XMT-1536 into registration-enabling studies based on its observed antitumor activity and favorable safety profile in an ovarian cancer population with a very poor prognosis and limited treatment options,” Protopapas added.

“Additionally, we look forward to presenting more comprehensive and mature results from the ongoing expansion cohort around the end of this year,” she concluded.

Clinical trials
First-in-Human Study of XMT-1536 in Cancers Likely to Express NaPi2b – NCT03319628

This article was published by Onco’Zine.

Powerful Together: The Clearity Foundation Recognized as Global Ovarian Cancer Charter Champion

The World Ovarian Cancer Coalition has designated The Clearity Foundation as a Champion of the newly launched Global Ovarian Cancer Charter which seeks to improve survival and quality of life for women diagnosed with ovarian cancer through the implementation of six Global Goals.  The Charter builds on the Coalition’s landmark 2018 Every Woman Study™, and was formally launched at the Presidential Plenary of the International Gynecologic Cancer Society xDigital Annual Global Meeting on 12 September 2020.

The Clearity Foundation has been recognized as a Charter Champion for its commitment to the Goals of the Charter, and because it already is starting new important work that speaks clearly to one or more of the Global Goals, that  has the potential to inspire others. A full list of Charter Champions can be found on the Coalition’s website; worldovariancancercoalition.org.

The Clearity Foundation is recognized for the following goals; Best Possible Care with our Treatment Decision Support Program and Information and Support with our Steps Through OC Program.

Learn more about the Champion Projects here!

“The Clearity Foundation firmly believes that women with ovarian cancer in the United States deserve the best possible therapies for their unique condition, and we are there to support them as they make treatment decisions. We are proud to support the Global Ovarian Cancer Charter as a Champion, particularly around access to best possible care and clinical trials.”

Worldwide, incidence of ovarian cancer is set to increase by almost 50% by the year 2050 and remains the most lethal of female cancers. While significant advances in treatment have been made in very recent years, women’s access to them is far from universal, especially in low- to middle-income settings, where the majority of women with ovarian cancer live.

Using the Charter as a framework for change, the Coalition is working with Charter Champions around the world to highlight inspirational projects or initiatives that may also foster global collaboration and sharing of good practice. Champions are mainly, but not limited to, patient and clinical organizations who have existing or upcoming programmes that speak directly to one or more of the Global Goals.  The aim is to reduce variations between regions and countries and ensure that women with ovarian cancer have the best chance of survival and best quality of life, wherever they may live.

“We are thrilled to be able to showcase such a diversity of Charter Champions. ” said Elisabeth Baugh, chair of the Coalition and CEO of Ovarian Cancer Canada, “We hope that their important work serves as an inspiration to collaborate and generate projects that speak to the Global Goals and, ultimately, to change the future of ovarian cancer for all women, no matter where they live.”

###

About Ovarian Cancer:

Ovarian cancer is the most lethal of the female cancers. Most women are diagnosed once the cancer has already spread, making it more difficult to treat. There are often delays in diagnosing the disease, as there is no early detection test, and symptoms are often confused with other, less severe, illnesses. Ovarian cancer is overlooked and underfunded – yet every woman is at risk of developing the disease. Ovarian cancer is cancer arising from the cells in and around the ovaries and fallopian tubes. It forms when there are abnormalities in normal ovarian cell growth. New cells grow unnecessarily, and old and damaged cells fail to die away, causing a build-up that forms a tumour or growth. Those growths have genetic mutations that cause them to multiply.

About the World Ovarian Cancer Coalition:

The World Ovarian Cancer Coalition is a not-for-profit organization that seeks to create a world where every woman with ovarian cancer has the best chance of survival, and the best quality of life – wherever she may live. With a stellar international Board of Directors, the Coalition builds on the success of World Ovarian Cancer Day (May 8) every year and the Every Woman Study as it looks ahead to further research, awareness raising, and policy work.

PARP Inhibitors Deemed Too Costly For Across-the-Board Frontline Maintenance in Ovarian Cancer

By Leah Lawrence

A new study estimated that a PARP inhibitor-for-all approach to treating advanced-stage ovarian cancer would be associated with high costs compared with biomarker-directed use of PARP inhibitors.1 Study researchers recommended that maintenance treatment with PARP inhibitors be reserved for patients with germline or somatic homologous recombination deficiency (HRD) mutations.

The study evaluated the cost of the 2 approaches by simulating the study designs of the PRIMA, VELIA, and PAOLA-1 trials. PARP inhibitors-for-all provided a greater progression-free survival benefit than biomarker-directed care for each trial, but was more costly as well.

The average cost per patients for the PARP inhibitors-for-all strategy was $166,269 for the PRIMA model, $286,715 for VELIA, and $366,506 for the PAOLA-1 model.

“The ranges in cost were dependent on the specifics of each of the 3 trials modeled including efficacy, timing of initiation of maintenance therapy (with initiation of standard chemotherapy or following completion of chemotherapy), presence or absence of bevacizumab in the baseline regimen, and cost of individual PARP inhibitors,” the researchers noted.

In comparison, when PARP inhibitors were given with a biomarker-directed strategy, the average cost per patient decreased to $98,188 for PRIMA, $167,334 for VELIA, and $260,671 for PAOLA-1.

The researchers also looked at the incremental cost-effectiveness ratios (ICERs) per quality-adjusted progression-free life-year (QA-PFY). PARP inhibitors-for-all frontline maintenance strategy was not cost-effective compared with a biomarker-directed approach, the researchers found.

Using an approach where every patient is treated compared with biomarker-directed treatment, ICERs were $593,250 per QA-PFY for PRIMA, more than $1.5 million per QA-PFY for VELIA, and more than $3.3 million per QA-PFY for PAOLA-1.

“Broadly speaking, a biomarker-directed approach that preferentially provides maintenance PARP inhibitor therapy for biomarker-positive patients might represent a more sensible value-based treatment model that exploits our understanding of ovarian cancer biology,” the researchers wrote. “This approach would not necessarily preclude biomarker-negative patients from receiving PARP inhibitor maintenance therapy but would encourage oncology providers to have individualized discussions with patients about costs, side effects, and expected benefits of their cancer treatment.”

Reference
Gonzalez R, Havrilesky LJ, Myers ER, et al. Cost-effectiveness analysis comparing “PARP inhibitors-for-all” to the biomarker-directed use of PARP inhibitor maintenance therapy for newly diagnosed advanced stage ovarian cancerGyn Oncol. Published online August 27, 2020. doi:10.1016/j.ygyno.2020.08.003.

This article was published by Cancer Therapy Advisor.

Patient-Reported Outcomes for Rucaparib as a Maintenance Therapy in Recurrent Ovarian Cancer

By Susan Moench, PhD, PA-C

Results of an exploratory analysis showed longer quality-adjusted progression-free survival (QA-PFS) and quality-adjusted time without symptoms or toxicity (Q-TWiST) in women with recurrent ovarian cancer treated with maintenance use of the poly(ADP-ribose) polymerase (PARP) inhibitor, rucaparib, compared with placebo as in the ARIEL3 trial. These findings were reported in the Journal of Clinical Oncology.

The phase 3 ARIEL3 study (ClinicalTrials.gov Identifier: NCT01968213)[SM1] , in which women with recurrent high-grade serous or endometrioid epithelial ovarian, primary peritoneal, or fallopian tube cancer who had achieved a complete response or partial response to platinum-based therapy were randomly assigned in a 2:1 ratio to receive rucaparib or placebo as maintenance therapy. Patients were stratified according the presence of a deleterious BRCA1/2 mutation, a non-BRCA alteration associated with deficient homologous recombination (HR), and no BRCA1/2 mutation with no evidence HR deficiency.

Results of the ARIEL3 study have been previously reported and showed significantly longer median progression-free survival (PFS), the primary study endpoint, in the intent-to-treat (ITT) population of patients receiving rucaparib maintenance therapy compared with placebo. Furthermore, while the most pronounced benefit was observed in those with a deleterious BRCA1/2 mutation, significant PFS advantages of rucaparib maintenance therapy were also observed in subgroups of patients with BRCA wild-type genes with or without evidence of HR deficiency.

Nevertheless, because most women diagnosed with ovarian cancer will experience disease relapse necessitating the administration of additional treatments that may be associated with significant toxicity, patient-reported outcome measures, in addition to assessments of PFS and overall survival, are particularly important endpoints of studies evaluating new treatments in this setting. In this context, safety assessments of patients enrolled in the ARIEL3 trial showed that while rucaparib was well tolerated, the frequency of grade 3 or higher treatment-emergent adverse events (TEAEs) was approximately 3.7-fold higher in the rucaparib compared with the placebo arm.

The focus of this post-hoc analysis of data from the ARIEL3 study was on patient-centered outcome assessments derived from the EQ-4D-3L questionnaire, which was completed on day 1 of each 4-week treatment cycle, at treatment discontinuation, and at a follow-up visit.

Two measures that incorporate assessments of both quality and quantity of life — the QA-PFS and the quality-adjusted time without symptoms or toxicity (Q-TWiST) — were determined for each patient.

Specifically, QA-PFS represents an adjustment to PFS that incorporates patient-reported toxicity and other detrimental effects of treatment, and was determined by multiplying the PFS function and the EQ-5D-3L index score function.

TWiST was determined by subtracting the mean time with grade 3 or higher TEAEs from the mean PFS, with Q-TWiST weighted according to patient-reported assessments of quality of life (QoL).

A comparison of the 2 treatment groups in the ITT population showed that QA-PFS was more than 2-fold higher for those receiving rucaparib compared with placebo. This significant difference was more pronounced in the subgroup with a BRCA1/2 alteration, where QA-PFS was 2.4-fold higher for those receiving rucaparib vs placebo, although a similar comparison showed a 1.6-fold difference for the subgroup without a deleterious BRCA1/2 alteration or evidence of an HR deficiency. Similar benefits of rucaparib over placebo were also reflected in the Q-TWiST endpoint.

“The Q-TWiST results also indicate that rucaparib maintenance treatment extended the time in which patients had good health status or QoL without cancer-related symptoms, which is a key objective for patients,” the study authors noted.

In their concluding remarks, the study authors noted that “by evaluating quality-adjusted survival, which incorporates assessments of quality and quantity of life, we demonstrated that rucaparib maintenance treatment provided significant benefit despite the impact of toxicities on patients’ health status during rucaparib treatment and that patients receiving rucaparib had longer periods without clinically relevant symptoms.”

This article was published by Cancer Therapy Advisor.